Skip to main content

Full text of "USPTO Patents Application 10681421"

See other formats


P50438-1 



Ma7I™R0MBOTIC 



JklSEHTS 



gTRU OF '™« TUVBSHm 

This invention relates to monoclonal antibodies 
,^s) that bind to a human coagulation - 
cofactor and their use as self -limiting rnhrbxtors of 
throKtoosls in combination with plasminogen activators. 

„,.^^^rs,Tm^ OF TP° TTOVEMTION 

under normal circumstances, an injury, be it mxnor 
or major, to vascular endothelial cells lining a blood 
Vessel triggers a hemostatic response through a se<^ance 
of events coi^only referred to as the coagulatron 
..cascade.- The cascade culminates in the conversion of 
sollle fibrinogen to insoluble fibrin which, -9-*^- 
Tith platelets, forms a localized clot or thro::tous which 
prevents extravasation of blood components. Wound 
Laling can then occur followed by clot dissolution and 
restoration of blood vessel integrity and flow. 

The events which occur between injury and clot 
f oration are a carefully regulated and linked series of 
reactions. In brief, a nu-toer of plasma coagulation 
proteins in inactive proenzyme forms and cofactors 
circulate in the blood. Active enzyme complexes are 
assembled at an injury site and are sequentially 
activated to serine proteases, with each successive 
serine protease catalyzing the subse^ent proenzyme to 
;rotease activation. This enzymatic cascade resul s in 
each step magnifying the effect of the ^^^^ 
For an overview of the coagulation cascade see the first 
chapter of ..Ttoombosis and Hemorrhage... J. Loscalzo and 
A, schafer. eds., Blackwell scientific Publications, 

Oxford, England (1994). M ^,,d 

While efficient clotting limits the loss of blood 
at an injury site, inappropriate formation of ^^o^ - 
veins or arteries is a con^on cause of disability and 



1 



P50438-1 



formation of clots on ^-^^^^f ^//J/,,,,, j^^art 
organs, shunts and prostheses such as artxfrcxa 

parenteral y . ^ • „„ „f the thrombin inhibitor, 

of clotting by activation of the th 
antithrombin III and inactivation of all 



factors 



However, due to their potency, heparin and L« 
patients receiving continuous 

vivo clotting txmes to be J" the 

contributes substantially to the cost of 
patient's inconvenience. ^^^.^^^ 
Further, the therapeutic target rang 

. ^ 1 .oi of efficacy without placing the 



P50438-1 



and at concentrations 
exceeds --- -- ana 

..eater than 4 "^'^^' fJ^/ZJ^,,, taken to Iceep the 

:rtre:r;. .rrrt^n. «... t. t„t. 

range. ^^,,iant with slower and 

Another approved anticoagulant wxt 

• effect is warfarin, a coumarxn 
longer lastxng effect xs ^^.^^ ^^.^ vitaxain K 

derivative. Warfarxn acts by ^ prothrombin 
dependent Post-translational .od f x^c^^^^^ 

and other Vitamin ^-^^P^^^^^'^/^;';,,,!, which blood 

=1 oattern of anticoagulant actxon, 
r;e:aerernon-c.otta..e at =oncent«-ons on^^^^^ 

-----T.rr.:rrrrLpa.n. 
"-^Tar/rcLa.. ----^--ra^;::a 

sustained reperfus.on of the .n „ 

rxrr:"r.T.-«.:.-.".- 
.«»""-rr^:irrrr..r"."„ ...... - 

unfractionated heparin, lo ^^^elet agents such 

airect throK^in inhibitors or ^^^^^^^^^^ 

■ • T^latelet glycoprotexn IlD/xxx 

as asp.r.n or platel g ^^^^^^ ^^.^ 

see Topol, ^ Heart J, 136, observation 
combination of therapxes is base 

..at Clot formation and a--^;-- "^^f^^^^^ ,..,,„ue 
processes and thrombin ^^^Z::^; and during 

a£ter the formation °* J <,,„ger et al, J 

„d after ---/--V;,. 7, LsB, . 
- coll Cardiol 31, «J ^^^^^^^^ 

...rers^e^ra:auabieage.s.d.^^^^^^^^^^ 

— — :"f:^f^^^ 



P50438-1 



York (1997)) s'^d thrombin activity e=chibits a 

:::::;;rJr:::e'.oiio.in. cessation o. ™ 

„,.b an ob.er.ea increase : 1,,.. 

hours following discontinuation of heparin. 

et a . Cat.e.erizatio. and Cardiovascular 

^4 25;-264 ,1998, and Granger, Circulation, 91 1929 

19;5 ,1995) . Further, antiplatelet agents may be 

■ A y.^r -bleeding or thrombocytopenia, 
accompanxed by bleedxng ^^^^ ^.^^ 

Also, numerous clxnical trxais nav 

aoses of thror^olytic agents lead to significant 

alteration in plasma hemostatic markers. See Rao et 

. J Clin invest, 101, 10-14 ,1988); Bovill et al.^ 

- T\ rsr-sfi — rir^ea^i: 

rceratirif .aa to —ed clot d^solution, 

the alteration in these hemostatic markers mirrors 
increased liabilities of thror^olytic therapy, 
;Lticularly the incidence of severe ble^ing^ 

Clearly, a need exists for antithrombotic agents 
efficacious in controlling thro:*otic disorders while 
maintaining hemostatic functions. 

fn»«.1.Y OF THVEHTIOM 

accordingly, one aspect of the present invention is 
a method for inhibiting thrombosis in an animal 
LIprising administering an effective dose of an anti- 
rolgulation factor monoclonal antibody having s If- 
limlting neutralizing activity in combination with 

plasminogen activator. „,,rhod of 

Another aspect of the invention is a method of 
reducing a retired dose of a thrombolytic agent in 
treatment of thrombosis in an , 
administering an anticoagulant ^^-^^^^^^^^^^J'^f ^'^^ 
component of the intrinsic coagulation pathway m 
combination with the thrombolytic agent. 

Another aspect of the invention is a method for 
reducing a recjuired dose of a thro^olytic agent 



4 



P50438-1 



treatment of thrombosis in an animal comprisxng 
administering an anti-Factor IX monoclonal ant.body xn 
combination with the thrombolytic agent 

pT,Tr.iP T^TTfirHTPTION »^ '^"^ DRAWINGS 

Figure 1 is a graph of experimental results 
demonstrating the titration of normal human plasma wxth 
the murine anti-Factor IX mAbs BCl and BC2 . 

Figure 2 is a graph of experimental results 
demonstrating the titration of normal human P^^-- -^^^ 
the murine anti-Factor IX mAbs 9E4(2)F4 and 11G4{1)B9. 

Figure 3 is a graph of experimental results 
demonstrating the titration of normal human plasma wxth 
the murine anti-Factor X mAbs HFXHC and HFXLC and the 
murine anti-Factor XI mAb HFXI. 

Figure 4 is a histogram of experimental results 
demonstrating the effect of heparin, acetylsalicylxc 
acid and murine Factor IX mabs on activated partial 
thromboplastin time (aPTT) at 60 minutes m a rat 
carotid thrombosis model. 

Figure 5 is a histogram of experimental results 
demonstrating the effect of heparin, acetylsalicylxc 
acid and murine Factor IX mabs on prothrombin txme at 60 
minutes in a rat carotid thrombosis model. 

Figure 6 is a histogram of experimental results 
demonstrating the effect of heparin, acetylsalicylxc 
acid and murine Factor IX mabs on occlusion of carotxd 
artery flow in a rat carotid thrombosis model. 

Figure 7 is a histogram of experimental results 
demonstrating the effect of heparin, acetylsalicylxc 
acid and murine Factor IX mabs on thrombus weight xn a 
rat carotid thrombosis model. 

Figure 8 is a histogram of experimental results 
demonstrating the effect of heparin, the murine Factor 
IX mab BC2, a chimeric Factor IX mab and humanxzed 



5 



P50438-1 



factor IX m^s on aPTT at 60 minutes in a rat carotid 

thrombosis model. 

Figure 9 is a histogram of experimental results 
demonstrating the effect of heparin, the -"-/^"^ 
ZX raab BC2 , a chimeric Factor IX ^ and hu.anr.ea 
factor IX mAbs on throrc^us weight rn a rat carotxd 
thrombosis model. 

Figure 10 is a histogram of experimental results 
demonstrating the effect of anti-Factor IX mab and 
heparin on tPA-mediated reperfusion. 

Figure II is a histogram of experimental results 
demonstrating the effect of anti-Factor IX mab and 
heparin on carotid vessel patency. 

Figure 12 is a histogram of experimental results 
demonstating the effect of anti-Factor IX mab and 
heparin on time to restoration of blood flow. 

Figure 13 demonstrates the effect of tPA on the 
hemostatic parameters, fibrinogen, plasminogen and 

^"'''Frg:r"e\4 demonstrates the effect of tPA, heparin 
and anti-Factor IX mab on aPTT. 

r.i.n.i.TT.ir.n DEF^^-^^t-rnw OF THF. INVENTION 

All publications, including but not limited to 
patents and patent applications, cited xn the 
fpecff ication are herein incorporated by reference as 
though fully set forth. 

The present invention provides a varrety of 
antibodies, altered antibodies and fragments thereof 
directed against coagulation factors, «hrch are 
Characterized by self-limiting 

preferably, the coagulation factor rs from ^he rntrrnsrc 
or coKo^on coagulation pathway. Most P-^"^^^;;*^^ 
anti-coagulation factor antibodies are ^'^---"^J^' 
anti-Factor IXa, anti-Factor X, 

Factor XI, anti-Factor XIa, anti-Factor VIII. antr 



6 



P50438-1 



Pactorvxxxa, anti-.actor V, anti-.actor Va ant.-.actor 
VII, anti-Factor Vila, anti-thron0.in or antx- 
prothrornJ^in. Particnlarly preferred are ant.-Factor 
antibodies. Exemplary anti-coagulation ^-"-^ 

. ■v,„,^ies are the humanized monoclonal antibodxes SB 

SB 257;32 directed against human Factor XX, the chrmer.c 
monoclonal antibody chaFIK directed 

IX the murine monoclonal antibodies BCX, BC2 , 9^*'2)^4 
and 1X04,1,B9 which are directed against ^ ° ^ 

.H/or Factor XXa or the murine monoclonal antibodies 

ain:xx which are directed against human Factors 

X and XI, respectively. Particularly P- ^"^^ 
anti-human Factor IX monoclonal antibody SB 2494X7^ 

The antibodies of the present invention can be 
prepared by conventional hybridoma techniques phage 
display coLinatorial libraries, i^unoglobulin chain 
t^ffUng and humani.ation techniques to generate novel 
self-limiting neutralizing antibodies. Also inc uded 
Ire fully human having self-limiting — » 

activity These products are useful in '^--P-"= 
Tharmaceutical compositions for thrombotic and embolic 
disorders associated with myocardial infarction, 
!: table angina, atrial fibrillation, strojce, renal 
damage, pulmonary e.*olism, deep vein thrombosis, 
percutaneous translumenal coronary angioplasty, 
disseminated intravascular coagulation, sepsis, 
artificial organs, shunts or prostheses. 

AS used herein, the term "self -limiting 

► refers to the activity of an 

neutralizing activity relers t 

antibody that binds to a human coagulation factor, 
preferably from the intrinsic and common pathways, 
rduding^actor XX/XXa, X/Xa, Xl/XXa VXIX/VXIIa a d 
v/va VXX/VIIa and thrombin/prothrombin and inhibits 
:hro;a.osis in a manner such that limited modulation of 
coagulation is produced. "Limited modulation of 



P50438-1 



coagulation" i. defined as an 

as measured by prolongation of the activated partial 
thromboplastin time (aPTT) , where plasma remains 
clottable with aPTT reaching a maximal value de-P^*^^ 
increasing concentrations of monoclonal antibody Th.s 
limited modulation of coagulation is in contrast to 
plasma being rendered unclottable and exhibitrng an 
infinite aPTT in the presence of increasing 
concentrations of heparin. Preferably, the maxrmal aP^ 
value of the methods of the invention are wxthrn the 
heparin therapeutic range. Most preferably, maxrmal 
aPTT is within the range of about 3 5 seconds to about 
100 seconds which corresponds to about 1.5 times to 
about 3.5 times the normal control aPTT value^ In one 
embodiment of the invention, aPTT is prolonged without 
significant prolongation of prothrombin time (PT) . 

The phrase "in combination with" refers to 
administration of one therapeutic agent before, after or 
concurrent with the administration of another 
therapeutic agent in a single course of treatment^ 

"Altered antibody" refers to a protein encoded by 
an altered in^unoglobulin coding region, which may be 
obtained by expression in a selected host cell. Such 
altered antibodies are engineered antibodies (e.g.. 
chimeric or humanized antibodies) or antibody fragments 
lacking all or part of an immunoglobulin 
region, e.g., Fv, Fab, Fab- or F(ab')2 and the liKa^ 

"Altered immunoglobulin coding region" refers 
nucleic acid secjience encoding an altered antibody of 
the invention. When the altered antibody is a CDR- 
.rafted or humanized antibody, the sequences that encode 
the complementarity determining regions (CDRs) from a 
non-human immunoglobulin are inserted into a first 
immunoglobulin partner comprising human variable 
framework sequences. Optionally, the first 



8 



P50438-1 



i^unoglobuUn partner is operatively linked to a second 

immunoglobulin partner. „„^lslc 
..First immunoglobulin partner" refers to a nuclerc 
acid secjuence encoding a human framework or human 
i:™unoglobulin variable region in which the - 
naturally-occurring, CDK-encoding -^^^ ^^^^f 
by the CDR-encoding regions of a donor antxbody 
hLan variable region can be an immunoglobulrn heavy 
Chain, a light chain (or both chains,, an analog or 
functional fragments thereof. Such CDR regxons, located 
within the variable region of antibodies 
(immunoglobulins, can be determined by Known methods rn 
the art. For example Kabat et al . in "Se^ences of 
proteins of Immunological Interest-, 4th Ed., U.S. 
Department of Health and Human Services, ^^^^^^f 
institutes of Health (1987, disclose rules for locating 
CDRS in addition, computer programs are known whxch 
are useful for identifying CDR regions/structures. 

■■second immunoglobulin partner- refers to another 
nucleotide se^ence encoding a protein or 
which the first immunoglobulin partner xs fused .n frame 
L by means of an optional conventional linker sequence 
(i e , operatively linked, . Preferably, it is an 
Lunoglobulin gene. The second -'^-^f^/^fj;: 
may include a nucleic acid sequence encoding the entire 
constant region for the same (I.e., homologous, where 
the first a!d second altered antibodies are derived from 
the same source, or an additional ,i e., 

antibody of interest. It may be an — ^"^^^^"^f^^^ 
Chain or light chain (or both chains as part of a single 
polypeptide, . The second immunoglobulin partner is not 
limfted to a particular immunoglobulin class or isotype. 
in addition, the second immunoglobulin partner may 
comprise part of an immunoglobulin constant region such 
as found in a Fab, or F(ab„ (i.e., a discrete part of 
an appropriate human constant region or framework 



9 



P50438-1 



region) . Such second Inununoglobulin partner may also 

comprise a sequence encoding an integral 

proLin exposed on the outer surface of a host cell 

e g , as part of a phage display library, or a sequence 

encoding a protein for analytical or diagnostic 

detection, e.g.. horseradish peroxidase, P- 

^"^^rfe::; :::-.c, ... .a. .a^. or ..a.,, are used 
with their standard meanings. See, e.g., Harlow et al . 
in "Antibodies: A Laboratory Manual", Cold Sprrng 

Harbor Laboratory, (1988). ^^^^ribes 
AS used herein, an "engineered antibody describes 
a type of altered antibody, i.e., a full-length 
synthetic antibody (e.g., a chimeric or h-anrzed 
al^ibody as opposed to an antibody fragment) in which a 
Tortion Of the light and/or heavy chain variable domains 
of a selected acceptor antibody are replaced by 
analogous parts from one or more donor antibodies which 

Uificity for the selected epitope. 
such molecules may include antibodies characterized by a 
humanized heavy chain associated with an --"^^^"^ 
light chain (or chimeric light chain) , or vice versa. 
Engineered antibodies may also be characterized by 
alteration of the nucleic acid se^ences encoding the 
acceptor antibody light and/or heavy variable domain 
framework regions in order to retain donor antibody 
binding specificity. These antibodies call 
replacement of one or more CDRs (preferably all from 
the acceptor antibody with CDRs from a donor antibody 
described herein. 

A "Chimeric antibody" refers to a type of 
engineered antibody which contains a naturally-occurring 
Triable region (light chain and heavy ^--^"^ 
from a donor antibody in association with light and 
heavy chain constant regions derived from an acceptor 
antibody. 



P50438-1 



A ..huraanized antibody- refers to - type of 
engineered antibody having its CDRs derived fro. a non 
hun,an donor iwaunoglobulin, the remaining 
i^aunoglobuiin-derived parts of the - ^^^^^ 
derived from one or more human x^nunoglobul.ns In 
addition, frame^or. s.pport id.es may b^^^^^^^^^^^^^ 

ro:rt;rarsrr"a. ro:..^. ..s., ^odgson 

at al Bio/Technology, S, 421 (1991) ■ , , 

T;e term "donor antibody- refers to a monoclonal or 
recoIinaL antibody which contributes the nucleic ac.d 
se^ences of its variable regions CPKs or other 
functional fragments or ^l^^;- TZ.. 
immunoglobulin partner, so as to provi 
lunoglobuXin coding region and --"^-^--^^f 
altered antibody with the antigenic spec.fic.ty 
rtralizing activity ufeTn this 

antibody. One donor -^^-::^ZZZ:. 
invention is a mur.ne ^^""^^^^^^"^ ^^^^^ 3,i«ble 
monoclonal antibody designated as ^« ' 
donor antibodies include the -"-/^"''^"^'^f 3,, 
neutralizing monoclonal antibodies designated as BCl. 
9.4(2,.4, 11G4(1)B9, ^F^LC and HPXX^ ^^^^^^^^ 
The term -acceptor antibody reters t 
.ecor^inant antibodies heterologous to the donor 

partner. Preferably, a human antibody xs the accept 
antibody. 

"CDRs- are defined as the complementarity 
determfnLg region amino acid sec^ences of an antibody 
::::: are the hypervanable regions of i:™^og obulin 
,eavy and light chains. ' ."^^f 'J /j,! 

■■sequences of Proteins of Immunological Interest , 



P50438-1 



H(=alth and Human Services, 
Fd US. Department of Heaicn duv^ ^vv-oo 
f H^^lth (1987). There are three 
national ----^ ^ ^f^rllin COKs c. C.K .e.ions in 

ruled h^ein refers .= alX three heavy chain CBRs. or 
aXX tLee li.ht chain CDKs or hoth all heavy and all 
light Chain CDRs, if approprxate^ 

CDRS provide the majorrty of contact resrdu 

- — — rrtri:: rernredror- 

Tor-Lro^tairheavy and li.ht -in se^ence. 

and inci.de ^ rarrorrtriire^reTnt::::' 

::r;:re::ri:;rnLor ne.trali.in. a^iUty as the 
donor antibody fro. which they were 

By "Sharing the antigen binding spec f.c.ty or 
neutralizing ability" is :»eant, for --^^^ ^^^^ 
although BC2 ^y be characterized ^ ^ ^ 
of self-limiting neutralizing act.vrty, a CDR encode 
I nucleic acid sequence of BC2 in an appropriate 

tructural environment ^y have ^fj, 
activity. It is expected that CDRs of BC2 xn such 
en'rolents will nevertheless recognize the same 
epitope (s) as BC2 ^^^^ ^.^^^^ 

A "functional fragment xs a pa 

• a;:,TnP antiqen binding 

region) which retaxns the same -^^^S 

specificity and/or neutralxzxng abxlxty 
from which the fragment was derxved. ..^^ ^y 

An "analog" is an amino acid se<iuence modxfxed by 
at le"t one alino acid, wherein said modificatxon can 
: Chemical or a substitution or a -arrangement of a 
few amino acids (i.e., no more than 10,, whxch 
modification permits the amino acid ----- - 

the biological characteristics, e.g., antxgen 



12 



P50438-1 



^ affinitv, of the unmodified 

soecificity and hxgh attinity/ 

sequence. Exemplary analogs include silent »utat.ons 
which can be constructed, via substitutions, to create 
certain endonuclease restriction sites within or 
surrounding CDR-encoding regions. 

analogs may also arise as allelic variations . An 
..allelic variation or modification" is an alteration m 
the nucleic acid sequence encoding the amino acid or 
peptide sequences of the invention. Such variations or 
Ldifications may be due to degeneracy in the genetic 
code or may be deliberately engineered to provide 
desired characteristics. These variations or 
modifications may or may not result in alterations in 
any encoded amino acid sequence. 

The term -effector agents" refers to non-protem 
carrier molecules to which the altered .nti.boa.es 
and/or natural or synthetic light or heavy ^^^ams of the 
donor antibody or other fragments of the donor antibody 
^y be associated by conventional means. Such non- 
pritein carriers can include conventional carriers used 
m the diagnostic field, e.fl-., polystyrene or other 
plastic beads, polysaccharides, e.g., as used m the 
BIAcore (Pharmacia) system, or other non-protein 
substances useful in the medical field and safe for 
administration to humans and animals. Other 
agents may include a macrocycle, for chelating a heavy 
metal atom or radioisotopes. Such effector agents may 
Tso be useful to increase the half-life of the altered 
antibodies, e.ff., polyethylene glycol. 

For use in constructing the antibodies, altered 
antibodies and fragments of this invention, a non-human 
species such as bovine, ovine, monkey, chicken, rodent 
,e g murine and rat) may be employed to generate a 
desirable ir^unoglobulin upon presentment with a human 
coagulation factor . preferably factor IX/IXa. X/Xa. 
Xl/Xla. Vlll/Vllla, v/va, Vll/VIIa or 



13 



P50438-1 



throntoin/prothrorrtoln or a peptide epitope therefrom, 
conventional hybridoma techniques are employed to 
provide a hybridoma cell line secreting a non-human mAb 
to the respective coagulation factor. Such hybridomas 
are then screened for binding using Factor IX/IXa, X/Xa, 
Xl/Xia, Vlll/VIIIa, V/Va, Vll/VIIa or 
thrombin/prothrombin coated to 96-«ell plates, as 
described in the Examples section, or alternatively ».th 
biotinylated Factor IX/IXa, x/xa, Xl/XIa, Vlll/VIIIa, 
V/Va Vll/VIIa or thrombin/prothrombin bound to a 
streptavidin-coated plate. Alternatively, fully huznan 
mAbs can be generated by techniques known to those 
skilled in the art and used in this invention. 

one exemplary, self-limiting neutralising mAb of 
this invention is mAb BC2, a murine antibody which can 
be used for the development of a chimeric or humanized 
molecule. The BC2 mAb is characterized by a self- 
limiting inhibitory activity on clotting time. As 
measured by the aPTT assay, the effect of the BC2 mAb on 
clot time exhibits a maximal value of about 100 seconds. 
The BC2 mAb also binds Factor IXa, inhibits Factor IX to 
IXa conversion and inhibits Factor IXa activity. 
Divalent metal cofactors are required for activity, with 
the mAb exhibiting a greater preference for Ca over 
Mn=- The observed IC„ in the aPTT assay is 
approximately 50 nM. The BC2 mAb exhibits a species 
cross-reactivity with rat and is of isotype IgG2a. 

Other desirable donor antibodies are the murine 
„^s, BCl, 9E4(2)F4 and 11G4(1)B9. These mAbs are 
characterized by a self -limiting inhibitory activity on 
clotting time. As measured by the aPTT assay, the 
effect of these mAbs on clot time exhibits a maximal 
value of about 90 to 100 seconds for 9E4(2)F4 and about 
80 seconds for 11G4(1)B9. The BCl mAb also binds Factor 
ixa, inhibits Factor IXa activity but does not inhibit 
Factor IX to IXa conversion. A metal cof actor is not 



14 



P50438-1 



re<iuired for its activity. The observed IC,. £or BCl in 
l7:Zr assay is approximately 3B nM. The BCl ^ rs 

" "refirher desirable donor antibody characterized 
by a self-limiting inhibitory activity on clotting txn,e 
Z the .urine ^ H.X.C. As measured by the aPTT assay, 
the effect of the HFXLC n,Ab on clot time exhxbxts a 
:::ill value of about BO to SO seconds. ^ 
binds Factor X light chain, and inhrb.ts Factor X/Xa 
activity. The observed IC,„ in the aPT^ assay rs 
approximately 20 nM. ^-i 

vet another desirable donor antibody characterized 
by a self-limiting inhibitory activity on clott.ng trme 

the murine m^, HFXX . As measured by -J 
the effect of the HFXI mab on clot time exhibits a 
:::i:ri vame of about lOO seconds. The H.X.C m^ binds 
Factor XI and inhibits Factor xl/XIa ^. 
Observed IC„ in the aPTT assay is ^^^^^^^ 
V«lile not intending to be bound to any particular 
theory regarding the mechanism of action, these mAbs 
appe- to regulate coagulation by a non-competitive or 
Illosteric mechanism whereby only partial inhibition is 

^^'":;:ts mvention is not limited to the use of the 
BCl, BC2, 9E4(2)F4, 11G4(1)B9, HFXLC, HFXI or their 

■ v,i^ (n ^ CDR) sequences. Any other 
hvpervariable {i.e., ^ut^i = si • ^ -k,, 

a^ropriate high-affinity antibodies characterized by a 
se!f-Umiting neutralizing activity and corresponding 

OHS may be substituted therefor. Xdentification of he 
donor antibody in the following description as BCl, BC2, 
9E4(2)F4, 11G4(1)B9, HFXLC or HFXI is made for 
illustration and simplicity of description only. 

The present invention also includes the use of Fab 
fragments or F(abM2 fragments derived from mAbs 
directed against the appropriate human coagulation 
factor or cof actor. These fragments are useful as 



15 



P50438-1 



agents having self -limiting neutralizing actxvxty 
against coagulation factors, preferably against Factor 
IX/lXa, x/xa, Xi/Xia, Vlll/VIIIa, V/Va, Vll/VIIa or 
thrombin/prothronOoin. A Fab fragment contains the 
entire light chain and amino terminal portion of the 
heavy chain. An F(ab')2 fragment is the fragment formed 
by two Fab fragments bound by disulfide bonds. The mAbs 
BCl BC2, 9E4(2)F4, 11G4(1)B9, HFXLC and HFXI and other 
similar high affinity antibodies, provide sources _ of Fab 
fragments and F(ab')2 fragments which can be obtained by 
conventional means, e.g., cleavage of the mAb with the 
appropriate proteolytic enzymes, papain and/or peps.n, 
or by recombinant methods. These Fab and F(ab')2 
fragments are useful themselves as therapeutic, 
prophylactic or diagnostic agents, and as donors of 
sequences including the variable regions and CDR 
sequences useful in the formation of recombinant or 
humanized antibodies as described herein. 

The Fab and F(ab')2 fragments can be constructed 
via a combinatorial phage library (see, e.g.. Winter et 
al , Ann Rev Irmunol, 12,433-455 (1994)) or via 
immunoglobulin chain shuffling (see, e.g., Marks et al., 
Bio/Technology, 10. 779-783 (1992), which are both 
hereby incorporated by reference in their entirety, 
wherein the Fd or Vh immunoglobulin from a selected 
antibody (e.g., BC2) is allowed to associate with a 
repertoire of light chain immunoglobulins, Vl (or Vk) , to 
form novel Fabs . Conversely, the light chain 
immunoglobulin from a selected antibody may be allowed 
to associate with a repertoire of heavy chain 
immunoglobulins, vh (or Fd) , to form novel Fabs. Self- 
limiting neutralizing Factor IX Fabs can be obtained by 
allowing the Fd of mAb BC2 to associate with a 
repertoire of light chain immunoglobulins. Hence, one 
is able to recover neutralizing Fabs with unique 



16 



P50438-1 



3e<^ences (nucleotiae ana a:.ino acia, fro. *e chain 

"""rrBro:-o..e. .n...o...s ..s.r.... a.ove .a. 

The mAD variable heavy ana/or 

contribute ..^^worlc se^ences, CDR 

rerrrroL-^ente, a— 

-^^''^ "tro;t:rni::rrro:rartra li^oaie. .hi.h 
rrhrac:::i:er:. l ^^..^^ --in. .eci^icit. o. 

the donor antibody. invention, or 

nucleic acia se<^ences encoding the cnHso ^^^^^^^^ 

"^^rLc litate insertion of ^uta.eni.ea COK ana/or 
which tacixitdLc -rf^aions can be 

- - — - i": > 
::n "s: :fr;r.:rrr. ... >. 

SEQ ID NOs: 8, 9 ana 10 ^^.^.es of the BC2 

... nucleic ana ^.no ac a e^^e^ ^ ^ 

-^^^^^^^^^ -o. thi. re.ion are listea 

code various coding sequences may be constru 

Toae the variable heavy ^^^^ Til a. 

sequences and CDR sequences of the xnve 



17 



P50438-1 



orCDRs can be .seS to produce altered a„txbod.es. e.^.. 
cLLric or humanized antibodies or other engxneered 
TZo^ies of this invention when operatively conO^rned 
with a second immunoglobulin partner. 

Xt Should be noted that in addition to --"^ 
.ucleic acid sequences encoding portions of the altered 
antibody and antibodies described here.n, °^^er such 
nucleic acid se<^ences are encompassed by the present 
invention, such as those complementary to ^^"^ 
CDR-encoding sequences or complementary to the modrf.ed 
hLn frameworlc regions surrounding the CDR-encod.ng 

useful DNA sequences include those sequences 
rch lybr!:::: under stringent hybridization conditions 

Cf^e T. Maniatis et al . , 

ho t-He DNA sequences. bee, 

!:o lecxar Cloning: . Laboratory Manual-, Cold Spr.ng 
Harbor Laboratory (1982,. PP- 387-389. An exa:^le 
one such stringent hybridization condition rs 
hybridization at 4XSSC at 65"C. followed by a washing in 
O.IXSSC at eS'C for one hour. ^1'-""^;^^^-. ^^^^^ 
exemplary stringent hybridization condition is 50% 

4XSSC at 42°C. preferably, these hybridizing 
formamide, 4XSSC ac v.. ^. -.^^ 

sequences are at least about 18 nucleotides m 



DNA S 



--ll^irld-i— Lrirmrieri::-can encode altered 
...ibodies Which include engineered anti^dies such a. 
Chimeric antibodies and humanized antibodies^ . ^^s.r^ 
altered i,»unoglobulin coding region -""-^^"'^ 
encoding regions that encode peptides having the antigen 
specificity of a Factor IX/IXa, X/Xa, KX/XIa, 
Viri/VIXIa, V/Va, VIX/VIIa or thro:rtoin/prothrombin 
antibody, preferably a high affinity ant body sue, a 
provided by the present invention, inserted into 
18 



P50438-1 



-^^v ^uch as a human framework or 
immunoglobulin partner sucn 

human immunoglobulin variable regxon^ 

preferably, the first immunoglobulin partner 

operatively linked to a second inonunoglobulin partner^ 

of interest, for example an Fc region. Second 
iLno.lo.uiin partners .ay also include se„ 
encoding anotl^er immunoglobulin to "''^^ ^^^^f 
heavy chain constant region is fused xn frame or by 
TZ of a linker sequence. Engineered antxbod.es 
Tircted against functional fragments -alog^ of 
coagulation factors may be designed to elrcxt enhanced 
binding with the same antibody. 

The second immunoglobulin partner may also be 
associated with effector agents as defined above 
trcluding non-protein carrier molecules, " f/^^^ 
second i^unoglobulin partner may be operatrvely Ixnked 

bv conventional means. ^^loVMilin 
Pusion or linkage between the second xmmunoglobulxn 
partners, e.g., antibody se<^ences, and the effector 
agent may be by any suitable means, e.g., by 
rrentional covalent or ionic bonds, protern fusxons, 
or hetero-bifunctional cross-linkers, e.g., 
carbodiimide, glutaraldehyde and the like. Such 
technigues are known in the art and are described rn 
conventional chemistry and biochemistry texts^ 

Additionally, conventional linker -^^^^ fj;*^^ 
simply provide for a desired amount of space between the 
simpj-y effector agent may 

second iimunoglobulin partner and ^^^^''""^ ^ 

— r":::re:-^^^^^^^^^^ 

- "^1^. id TeUces for the molecules of 
the invention may be modified by techniques known to 
those skilled in the art to enhance expression. 



19 



P50438-1 



A preferred altered antibody contains a variable 
heavy and/or light chain peptide or protein sec^ence 
having the antigen specificity of BC2, e.g., the Vh 

aid V. Chains. Still another desirable altered ant.body 
of this invention is characterized by the amino acrd 
sequence containing at least one. and preferably all of 
the CDRs of the variable region of the heavy and/or 
light chains of the murine antibody molecule BC2 w.th 
the rer,>aining secjuences being derived from a human 
source, or a functional fragment or analog thereof. 

in a further embodiment, the altered antibody of 
the invention may have attached to it an additional 
agent For example, recombinant DNA technology may be 
used to produce an altered antibody of the invention m 
which the FC fragment or CH2 CH3 domain of a complete 
antibody molecule has been replaced by an enzyme or 
other detectable molecule (i.e.. a polypeptide effector 
or reporter molecule) . 

The second immunoglobulin partner may also be 
operatively linked to a non- immunoglobulin peptide, 
protein or fragment thereof heterologous to the CDR- 
containing sequence having antigen specificity to a 
coagulation factor, preferably to Factor IX/IXa, X/Xa, 
Xl/Xia, Vlll/VIIIa, V/Va, Vll/VIIa or 

thror^in/prothrombin. The resulting protein ^^"^^^^^ 
both antigen specificity and characteristics of the non- 
i^nunoglobulin upon expression. That fusion P-"-' 
characteristic may be, e.g., a functional characteristic 
such as another binding or receptor domain or a 
therapeutic characteristic if the fusion partner is 
itself a therapeutic protein or additional antigenic 
characteristics. 

Another desirable protein of this invention may 
comprise a complete antibody molecule, having full 
length heavy and light chains or any discrete fragment 
thereof, such as the Fab or F{ab')2 fragments, a heavy 



20 



P50438-1 



chain dimer or any .ninimal recorrtoinant fragments thereof 
chain ^i^gi^.chain antibody (SCA) or any 

such as an Fv or a siiiy-^ cc.l*=rted 
other raolecule with the same specifxcrty ^^'^^ l^^^^'^^ 
donor r^. e.,., ^ BCl, BC2, 9K4,2,.4 l^-^^'^*-/;^ 
HFXLC or HFXI. such protein may be used in the form 
al altered antibody or may be used in its unf used form. 

vmenever the second immunoglobulin partner rs 
derived from an antibody different from the ^onor 
Ttibody, e.... any isotype or class of i^nunoglobulin 
™r. or constant regions, an engineered antrbody 
results. Engineered antibodies can comprise 
i^aunoglobulin ,Ig, constant regions and -^-^^^ 

=-r;r.rjr 

donor antibody, e.g., the anti-Factor ix/ixa, X/xa, 
Xl/Xia, Vlll/VIIIa, V/Va, Vll/VIIa or 
.hrombin/prothro^in antibodies described ^ 
^ ^ f^e^l e-tions , substitutions, 
addition, alterations, e.g., deletions, 
ridditions, of the acceptor ^ lig^t and.or heavy 
variable domain frameworK region at ^-."-1-; -/^J^ 
amino acid levels, or the donor CDR — -"^^f 
in order to retain donor antibody antigen binding 

^'^'^"suirengineered antibodies are designed to employ 
one (or both, of the variable heavy and/or light chains 
of the coagulation factor (optionally modified as 

CDRs. The engineered antibodies of the invention 
exhibit self-limiting neutralizing activity. 

such engineered antibodies may include a humanized 
antibody containing the frameworlc regions of a selected 

immunoglobulin or subtype or a chimeric antibody 
containing the human heavy and light chain constant 
regions fused to the coagulation factor antibody 
functional fragments. A suitable human (or other 
animal, acceptor antibody may be one selected from a 



21 



PS0438-1 



n ^.f^hase eg., the KABATO database, Los 
ccnventxonal ^.^.^.e,JJ - ^^^^^^^^ ^ ^^^^^ 

Alamos database, and Swiss fro 

to the nucleotide and amino acid sequences of the donor 
antibody A human antibody characterized by a homology 
: ::: Irame„or. regions o. the donor antibody <on^n 
amino acid basis, may be suitable - P-^J^J 
chain variable framework region for .nsertxon of the 
lonor CDRs. A suitable acceptor antibody capable of 
riing light Chain variable -^^mewor^^^^^^^^^^^^^ 

required to originate from the same acceptor 

preferably, the heterologous framework and constant 
regions are selected from human ^^^^^^^^ 
and isotypes. such as IgG (subtypes 1 through 4) , IgM. 

aTxgB. However, the acceptor antibody need not 
cimprise only human immunoglobulin protein ---^-^ 
Por instance, a gene may be constructed xn whxch a DNA 
^^Hlna part of a human immunoglobulin chain 
Ts^:::: ZTZ iZ^r... encod^g a non-i„Xobulin 
lli" acid sequence such as a polypeptide effector or 

reporter molecule. 

A particularly preferred humanized antibody 
contains CDRs of BC2 inserted onto the framework regions 
of a selected human antibody se^ence. For neutralizing 
iLized antibodies, one. two or P-^^- 
from the Factor IX antibody heavy chain and/or light 
Chain variable regions are inserted into the framework 



regions of the selected human antibody sequence, 
replacing the native CDRs of the latter 

preferably, in a humanized antibody, ^^^^J^"^^^^ 
aomains in both human heavy and light chains have been 
enaineered by one or more CDR replacements. It 
Tosslble to use all six CDRs, or various co.a.inations of 
les than the six CDRs. Preferably all six CDRs are 
replaced. It is possible to replace the CDRs only in 

22 



P50438-1 



the wan heavy chain, using as light chaxn the 
^^odified light Chain frc. the huraan acceptor antibody. 
Still alternatively, a compatible light cha.n :«ay be 

cted fro» another human antibody by ---- 
conventional antibody databases. The ^- "^^^'^^ 

engineered antibody may be derived from any suitable 
acceptor human immunoglobulin. 

The engineered humanized antibody thus preferably 
has the structure of a natural human antibody or a 
fragment thereof, and possesses the combination of 
pro^rties retired for effective therapeutic use^e. ^. . 
treatment of thrombotic and embolic diseases in man 

Most preferably, the humanized -tibodies have a 
heavy chain amino acid se^ence as set forth in SEQ ID 
To .1. 52, or 89. Also most preferred are humanized 
antibodies having a light chain amino acid se^ence as 
set forth in SEQ ID NO: 44, 57, 62, 74 or 99. 

particularly preferred is the ^---^^ 

249413 v,here the heavy chain has the ammo -"^-^J" 
as set forth in SEQ ID m-. 31 and the light chain has 
tL alino acid se.^ence as set forth in SBQ ID NO: 44. 
tl!o r-articularly preferred is the humanized antibody SB 
r4inrere t JhLvy Chain has the amino -id se^ence 
as set forth in SEQ ID NO: 52 and the light chain ha 
the amino acid se^ence as set forth in ^ J-^^^' 
Mso particularly preferred is the humanized antibody SB 

- :::: t:^ 

— tit:— ^^^^^ 
-rr::rn%r7D:-::d-^^^^^^^^^^ 

L amino acid sec^ence as set forth in ^^^^ 
Mso particularly preferred is the J^";;^^//, 
257731 where the heavy chain has the ammo 
as set forth in SEQ ID NO: 52 and the light cham has 



23 



P50438-1 



..e an,ino acid sequence as set fort^ ^^^^^f ^J^'^^ 
Also particularly preferred is the humanized --^^^'^^ 
' ,32 Where the heavy chain has the a»ino -^^--J" 
as set forth in SEQ ID NO: 89 and the light charn has 
:L a^ino acid sequence as set forth in SKC .0: 

It will be understood by those skilled -"^he art 
that an engineered antibody ™ay be further modrfred by 
Changes in variable domain a»ino acids without 
necessarily affecting the specificxty and h gh aff.nrty 
of the donor antibody (i.e., an analog). xs 
aLicipated that heavy and light chain amino acxds :»ay 
be substituted by other amino acids erther m the 
variable domain frameworks or CDRs or both. These 
substitutions could be supplied by the donor antxbody 
consensus se<^ences from a particular 

in addition, the constant region may be altered 
enhance or decrease selective properties °^ 
molecules of this invention. For example, 
binding to .c receptors, or the ability to bxnd nd 
activate complement (see, e.g., imgal et -i'' "° 
x™.unoI, 30, 105-108 ,I9«, , Xu et 

269 3469-3474 (1994), Winter et al., EP 307434-B). 

^ altered antibody which is a chimeric antxbody 
differs from the humanized antibodies described above by 
providing the entire non-human donor antibody heavy 
chain and light chain variable regions, xncludxng 
framework regions, in association with human 
i™„unoglobulin constant regions for both chaxns^ It 
anticipated that chimeric antibodies whxch retaxn 
additional non-huxnan se^ence relative to 
antibodies of this invention may elicit a sxgnxfxcant 
immune response in humans. 

such antibodies are useful in the preventxon and 
treatment of thrombotic and errbolic disorders, as 
discussed below. 



24 



P50438-1 



preferably, the variable light and/or heavy chain 
sequences and the CDRs of ^ BC2 or other 

e..., BCl. 9B4(2,.4, lia4<l,B9, 3. 
and their encoding nucleic acid se-juences. "^^^^^^ 
in the construction of altered antibodies, preferably 
humanized antibodies, of this invention, by the 
following process. The same or ^i-^^^^-^^^^^^^^ 
also be employed to generate other embodiments of th.s 

invention. av. o 

J A-^r^ a c;^l e^cted donor mAto, e.g., 
A hybridoma producxng a seieccea 

the murine antibody BC2, is conventionally cloned and 
the DNA of its heavy and light chain 

obtained by techni^es known to one of slcrll .n the art, 
e <, the techniques described in Saittrook et al., 
..M^llcular Cloning: A Laboratory Manual " , 2nd ed.t.on, 
cold spring Harbor Laboratory (1989) . The ---=1^ 
heavy and light regions of BC2 containing at least the 
CPR^ncoding regions and those portions of the acceptor 
mAb light and/or heavy variable domain framework regrons 
required in order to retain donor mAb bindrng 
specificity, as well as the remaining iirmunoglobul.n- 
Trived parts of the antibody chain derived from a human 
i^unoglobulin, are obtained using polynucleotrde 
primers and reverse transcriptase. The CDR-encod.ng 
regions are identified using a Icnown database and by 
comparison to other antibodies. 

A mouse/human chimeric antibody may then be 
prepared and assayed for binding ability. Such a 
chimeric antibody contains the entire non-human donor 
antibody V„ and Vl regions, in association with human Ig 
constant regions for both chains. 

Homologous framework regions of a heavy chain 
variable region from a human antibody are ^^^^^^^^ 
using computerized databases, e.^., KABA^ and a human 
antibody having homology to BC2 is selected as the 
acceptor antibody. The sequences of synthetic heavy 



25 



P50438-1 



Chain variable regions containing the BC2 CDR-encoa.ng 
regions within the human antibody frameworks are 
designed with optional nucleotide replacements rn the 
framework regions to incorporate restriction sxtes 
This designed sec^ence is then synthesized usrng long 
synthetic oligomers. Alternatively, the designed 
sequence can be synthesized by overlapping 
rifgonucleotides, amplified by P^-erase chain reaction 
(PCR). and corrected for errors. A suitable light chain 
variable framework region can be designed in a similar 

manner- , 

A humanized antibody may be derived from the 
Chimeric antibody, or preferably, made ^vnthetically by 
inserting the donor mAb CDR-encoding regions from the 
heavy and light chains appropriately within the selected 
heavy and light chain framework. Alternatively, a 
humanized antibody of the invention may be prepared 
using standard mutagenesis techniques. Thus, the 
resulting humanized antibody contains human framework 
r::ions and donor mAb CDK-encoding regions. .here may 
be subsequent manipulation of framework residues, 
resulting humanized antibody oan be expressed in 
reco^cO^inant host cells, e.g., COS, CHO or myeloma cells . 
Other humanized antibodies may be ^^-^-"^^l^.^^l^Hl^ 
technique on other suitable Factor IX-specific or other 
coagulation factor-specific, self-limiting, 
neutralizing, high affinity, non-human antibodies. 

A conventional expression vector or recombinant 
plasmid is produced by placing these -^^"^ --"^"^f^^ 
for the altered antibody in operative association with 
conventional regulatory control sequences "P^^^ f 
controlling the replication and expression m. and/or 
secretion from, a host cell. Regulatory sequences 
include promoter se,^ences, e.g., CMV promoter and 
signal sequences, which can be derived from other known 
Ttibodies. Similarly, a second expression vector can 



26 



P50438-1 



,e proaucea .avin. a DNA se^ence »l.ic. encode, a 
..„p.e.en.a.. ^^^^^^ ^^^^^ ^^^^^^"^ 

conventional technic^es w.th both the £ 

vectors ,or si»ply ---ff^^f , L invention 

create the transfected host ^^..^^i^ Ught and 

rrr,:: ":r:-. " " 

other altered antibodies and molecules of th.s 

'"^'"surtable vectors £or the cloning and subclonin. 
steps^ploved i. the .ethods and oonstruct.o h 
compositions Of this Of 

fro. ..... 

r—i; rrrpi:::!. readH. ha. a„ ^ndance Of 
not a limiting factor in this invention. 



27 



P50438-1 



Similarly, the vectors eT^loysd for expression o£ 
eUeere. a.ti.oaies oc r. t..^r^n..o„ 

- cfor" tIc Te rs also contain selected 

conventronal vector. The ^^^.^.^^ „hich 

regulatory sequences (such ^J^J j^^.^.^.o^ous 

airect the "-^^-"""^"/JJ/:" s. These vectors 
se^ences in selected 

efected i:™»unoglo.ulin se^ences -o^^^JJ 
insertion of desirable restriction srtes for ready 

manipulation. characterized by 

The expression vectors may also D 
genes suitable for amplifying expression of the 
^ ^ rr the maminalian 

l^eterologous DNA other preferable 

aihydrofolate -<^---;;";„ °T 3e.uence, such 

vector sequences rnclude a poly A g 

.s from bovine growth sectors 
nromoter sequence (betaglopro) . The exp 
Tslful here" may be synthesized by techniques well 
known to those skilled in this art. 

The components of such vectors, e.g.. replico , 

— — ^^^^^^^^^ 

r synthLi^ed by .nown p— 

airecting the expression and/or secretron of the P 
„f the recombinant DNA in a selected host. Other 

Z expression vectors of which numerous types 
appropriate expression K-cterial, insect, 

are known in the art for —^^^"'^f 
yeast and fungal expression may also be selecte 

TeTesent invention also encompasses a cell line 

^sfectL with 



28 



P50438-1 



.He Cloning and other manipulations of ^-^ J^-f/^^ 
vectors are also conventional. However, 

• ^ r^-f TT coll are usea tor 
rells from various strains of E. co±i a 
replication o. t.e cloning vectors an. other steps .n 

the construction of altered antibodxes of thrs 

invention. 

suitable host cells or cell Imes for the 
expression of the engineered antibody or altered 

of the invention are ---- ^ ; ^--^^^ 
such as CHO, COS, a fibroblast cell (e.g., 3T3) an 
ireloid cells, and .ore preferably a CHO or a .yelo.d 

Hu»a„ cells .ay be used, thus enabling tbe 
molecule to be modified with human -Ivcosylatxon 
patterns. Alternatively, other eukaryotxc cell Ixnes 

be employed. The selection of 
host cells and methods for transf ormatxon, culture 
rUfication, screening ^ -rr"s::^^oo. 
purification are known in the art. See, e.g , 

et al. , supra. 

Bacterial cells may prove useful as host cells 
.or the expre s. the reco—^^ 

of proteins expressed in bacterial cells to be xn an 
unfolded or improperly folded form or in a non- 
glycosylated form, any recoKtoinant Fab produced .n a 
fafterLl cell would have to be screened for retentron 
of antigen binding ability. If the molecule expressed 
t the bacterial cell was produced in a P-P"^- "^^^^ 

.Lm, that bacterial cell would be a ^-"^^^^ 

Por example, various strains of E. CO a 

expression are well-lcnown as host cells .n the fxeld 

biotechnology. Various strains of B. 

Streptomyces, other bacilli and the Ixke may also be 

employed. 



29 



P50438-1 



«>ere desired, strains of yeast cells )cnown to 
..ose skilled in t.e art are also available as .o^t 
cells, as well as insect cells, e... Prosoph. a and 
Leoidoptera and viral expression systems. See, eg. 
Lepiaopcexo 277-298, Pleniim 

Miller et al.. Genetic Engineering, 8, 277 

required to produce the host cells ^ 
culture methods necessary to produce the -"--^ 
antibody of the invention from such host cells are all 
conventional techni^es . Likewise, once -^<'^-'^,^,'J''^^^ 
altered antibodies of the invention may be purrfred from 
the cell culture contents according -^^-"^"^ 
procedures of the art, including ammonium sulfate 
preciSi-^i-. -«^-^^ chromatography. 
Tel electrophoresis and the liKe. Such techniques are 
Within the skill of the art and do not limit this 

'"^nlranother method of expression of the humanized 
antibodies may utilise egression — 3^,. 

animal, such as described m U. S. Patent 
This relates to an expression system using the animal s 
casein promoter which when transgenically --J- 
into a maiM^al permits the female to produce the desired 
recombinant protein in its milk. 

once expressed by the desired method, the 
engineered antibody is then examined for in vitro 
activity by use of an appropriate assay. Presently, 
conreLIonal K.XSA assay formats are employed to assess 
g:alitative and .^antitative binding of the engineered 
antibody to Factor IX or to other -P^-^"^^^ 
coagulation factors. Additionally, 

assays may also be used to verify neutralising efficacy 



30 



P50438-1 



evaluate the persistence of the engineered antxbody rn 
the body despite the usual clearance mechanxsms. 

Pollowin. the procedures described for hu^anrzed 
antibodies prepared from BC2 , one of skill in the art 
Tay all construct humanized antibodies fro„ — 
Iltibodies, variable region sec^ences and CDR peptrdes 
irs ribed herein. Engineered antibodies can be produced 
„ith variable region frameworks potentially recognrzed 
as -self by recipients of the engineered antibody 
Tinor modifications to the variable region ; 
can be implemented to effect large increases « -'J^ 
binding without appreciable increased immunogen.city 
the recipient. Such engineered antibodies may 
effectively treat a human for coagulation factor- 
mediated conditions. Such antibodies may also be useful 
in the diagnosis of such conditxons. 

This invention also relates to a method for 
inhibiting thro.4.osis in an animal, particularly a 
:Lan, Which comprises administering - effectrve dos 
of an anti-coagulation factor monoclonal antxbody havxng 
self-limiting neutralizing activity in co:*xnatxon wxth 
a plasminogen activator. Co:<^ination therapy enhances 
thrombolysis at sub-optimal 

plasminogen activator decreasing the txme to 
of blood flow and increasing the frequency and total 
duration of vessel reperfusion. In contrast to heparxn. 
cox,*ination therapy does not significantly perturb 
normal hemostatic functions and spares fxbrxnogen, 
plasminogen and alpha-2-antiplasmin levels. 
Accordingly, this invention also relates to a method 
reducing a re,^ired dose of a throTtoolytxc agent xn 
treatment of thrombosis in an animal comprxsxng 
adllnistering an anticoagulant specifically targetxng a 
component of the intrinsic coagulation pathway xn 
combination with the thrombolytic agent. 



31 



P50438-1 



preferably, the coagulation factor is from the 
intrinsic or co,«aon coagulation pathway. Most 
preferably, the anti-coagulation factor ■nonoclonal 
pretera y, anti-Factor Ixa, anti- 

antibody IS an anti-Factor i p.^tor 
Factor X, anti-Factor xa, anti-Factor XI, anti Factor 
X a anti-Factor VXXX, anti-Factor VXXIa, ----^J: 

Factor VII, anti-Factor Vila, anti 
rhlll:: or anti-prothro^^m. The ^ can include one 
or more of the engineered antibodies or altered 
antibodies described herein or fragments thereof. 
Preferably, the plasminogen activator is tPA, 

re:to.inase, urokinase or tPA variants as described 

— ---^-^r:r°";/rcrti r.s^ -B^r; 

14585 (1997); FUDlse et al . , Circuia 

,,997,, coombs et al., .Biol C.e™, ^"'/f ^^^^^^ 
,1998); van de Werf at al., A. Heart J, 137, 786 
,1999) . Particularly preferred is tPA. 

Alternatively, acetylsalicylic acid can be 
administered in con*ination with the anti-coagulation 
:rtor monoclonal antibody. Xn some cases, combine ion 
therapy lowers the therapeutically effective dose of 
anti-coagulation factor monoclonal antibody. 

L therapeutic response induced by the use of 
the molecules of this invention is produced by the 
binding to the respective coagulation factor t^^ 
s^blc^ent self-limiting inhibition of the coagulation 
cascade Thus, the molecules of the present invention, 
:Zln preparations and formulations appropriate for 
Therapeutic use, are highly desirable for person3 
susceptible to or experiencing abnormal clotting 
acti'ty associated with, but not limited to, myocardial 
infarction, unstable angina, atrial fibrillation, 
strolce, renal damage, pulmonary e^olism, deep vein 
:hromb;sis and artificial organ and prosthetic implants. 
A particularly preferred use is in myocardial 
infarction. 



32 



P50438-1 



The altered antibodies, antibodies and fragments 
thereof of this invention may also be used ^n 
on lotion with other antibodies, particularly h^n 
reactive with other markers (epitopes, responsible 
Tr the condition against which the engineered ant.body 
of the invention is directed. 

The therapeutic agents of this invention are 
believed to be desirable for treatment of abnormal 
Clotting conditions from about 1 day to about 3 weeks, 
or as needed. This represents a considerable advance 
over the currently used anticoagulants ^^^^^^ 
warfarin. The dose and duration of treatment relate, 
the relative duration of the molecules of the present 
inveltion in the human circulation, and can be adjusted 

Tone o* ^-^'^ ^^'^ ''^^^"^"'"^ 11 

being treated and the general health of the patxent. 

The mode of administration of the therapeutic 
agents of the invention may be any suitable --^e whrch 
Tllvers the agent to the host. The altered antrbod.es, 
antibodies, engineered antibodies, and fragments 
thereof, plasminogen activator and pharmaceutical 
colposi ions of the invention are particularly useful 
for parenteral administration, i.e., subcutaneously . 
intramuscularly, intravenously or intranasally^ 

Therapeutic agents of the invention may be prepared 
as pharmaceutical compositions containing an 
lount of the engineered .e.g.. humanized, antibody of 
the invention and plasminogen activator as active 

ngredients in a pharmaceutically acceptable carrier^ 
Alternatively, the pharmaceutical compositions of the 
invention could also contain acetylsalicylic acid, 
the prophylactic agent of the invention, an acraeous 
suspension or solution containing ^^^^^-^"^ ,^ ^ 
antibody, preferably buffered at physiological pH, in 
orl ready for injection is preferred. The compositions 
for parenteral administration will co,»only comprise a 



33 



P50438-1 



r^vc^f f:>rablv an aqueous carrier, 
acceptable carrxer, preferably ^ ^ o.4% 

variety of aqueous carriers may be employed eg , 
variety o^ These solutions are 

saline. O'^* ^^-^-^/"tro.'L.iculate -natter. T.ese 

sterile and ^^^^^/^^f^/J J,,„.entional, well Kno„„ 
solutions may be sterilizea oy 

.;„,.i.„i-. 

or 20* by -^^*'\-^^7;,^^;^::'^'^L.oLing to the 
fluid volumes, viscosities, etc., 

particular r.ode of -^'^'^';"''°\Z'Z l,\^^ invention 
^us, a Pharmaceutical ^^'^^^^^^'^jl^^ ,o„,,iu 

rLTtiretLreH^^^^^^^^ ^ - - 

rut^rorm. e... a^ut --^rl^rred 
preferably, about 5 m. to about 25 

.„.,.ody Of - rtion'f:: intrlvenous infusion 

composition of tne mve sterile 
..uld be made up to .SO ^o. . 

rfrra^^r" - » — : -^^'^-^^ 

preter ^ n^ethods for preparing 

of the invention. Accua are well known 

parenterally administrable -----^ "^^^ 

- wnl -.--ent to t, -1 led^^ 

rr^relLrrcire^^ ed., Mac. Publisbin. 

--Tt\r:re:errertrre-t.erapeutic a.ent. of tbe 
invention, w.en in a pbarmaceutical preparation, be 



P50438-1 



... .... .= """"r .... .. I p....- 

:r.;"i:*r.„„ .... - — • - 

employed . 

The present Invention will now be described with 
.e^erlnce to the following specific. non-l.n>.trna 
examples . 



35 



P50438-1 



..a scr g of >n.i-F...or TX Monoclonal . 

ani-i bodies 

Female Balb/C mice were injected with human factor 
IX purified as described in Jenny, R. et al.. Prep 
Bloche., 16. 227-245 (1986). Typically, each mouse 
received an initial injection of 100 ug P"^"" 
dissolved in 0.15 m. phosphate-buffered salrne ,PBS, and 
mixed with 0.15 m. complete Preund.s ^^^^^^^^ 
i^unizations of 50 ug protein in 0.15 mL PES w.th 0 15 
m. incomplete Preund-s adjuvant were given approximately 
biweelcly over a 2-3 month period. After the frnal 
boost, the mouse received 50 ug of Factor IX rn PBS 
three days before spleen/myeloma cell fusxons. Spleen 
cells were isolated from an immunized mouse and fused 
with NS-1 myeloma cells (Kohler, G. e£ al . , Bur J 
Immunol, 6. 292-295 (1976,) using polyethylene glycol as 
described by Oi, V.T. et al . in -Selected Methods .n 
cellular Immunology," Mishell, B.B. and Shigix, S.M.. 
eds . Freeman Press. San Francisco. Following the 
fusion, the cells were resuspended in RPMI 1640 med.a 
containing 10% fetal calf sera and alicjuots were placed 
in each well of four 24-well plates contarnrng 0.5 mL 
peritoneal lavage cell-conditioned media. On the ^ 
following day, each well received 1.0 m. o 2 x 10 
hypoxanthine. 8 x lO" M aminopterin and 3.2 x 10 M 
t^idine in RPMI 1640 media containing 10% fetal calf 
se^ The cells were fed every 3-4 days by removing 
half of the media and replacing it with fresh medra 
containing 1 x IQ- M hypoxanthine and 1 . 6 x 10 M 

thymidine. 1 0 mL of hybridoma 

Approximately two weeks later, i.u mu 
medium was removed from each well and tested for antr- 
Factor IX antibodies using an E.ISA assay as descrxbed 
by Jenny, R.J. at al . in Keti, Bmy^ol. 222. 400-416 
,1993). Briefly, factor IX was immmobilized onto 



36 



P50438-1 



11. of 96-well microtiter plates. Hybrxdoma 
plastxc wells of 96 wel antibody were then 

supernatants or dilutions of purxfxed 

substrate o-dianisidine^ ,„,ibodies were 

wells containing anti-Factor 
subcloned by limiting dilution and grown in 96 well 
nlates supernatant from the cloned hybridoma cell 
plates, bup antibody to Factor IX by the 

^„lt-iires were screened for antmouj 

Hssay described above and cells ^^^J^ 

.y^ridomas were — -^iZrul^s^ re 
nitrogen and then grown as ascitic 

, ,^ , ;^..-rn, . rna.ion Facto r 

The effect of increasing concentrations of anti- 
coagulation factor antibodies on activated partial 
thro^oplastin time (aPTT, of human plasma was 
aetermined in a fibrometer (Becton-DicHinson 
microbiology Systems, '^-^^^rT^Z:;^ 
Baxter reference procedure LIB0293 J, i/^ 
(Baxter Scientific, Edison, New Jersey) 

prior to the start of the experiment, 2 to 3 ml. 

. tube were placed into the heating 

n no M raCI in a 5 mL tuPe weie fi 

Of ;he ..rometer^ ^an Plasma sa^^^^^^^^^ 

reference Plasma (^^erican Diagnostics, Greenwich, 

^"""^rfirctionated heparin from porcine intestinal 
T ci- Tnnis Missouri), low 

:re::irrigrh:::i:f.rp:r;ine intestmai mucosa 

Cenox*, enoxaparin sodium. Rhone-Poulenc Korer 



P50438-1 



t-^rals Collegeville, Pennsylvania) or mAb 
Pharmaceuticals, Coixey« .^^t-elv BO uM 

anticoagulant was included as a ^^^^l^^^ ^.^^ ,oO 

TWO fibroTube* fibrometer cups were fiHea w 
.1 tert Plasma or 100 ul test plasma with anticoagulant 
L 125 ul of actin activated cephaloplastin reagent 
r^lirreagent. fro. rabbit brain ^---^ 
aoid, available from Baxter Screntrf xc) , ^respectw y 
and placed in the fibrometer wells at SVC. 

. 100 ul of actin reagent was 

after one minute, luu u± oi. a 
..ansreLd to a plasma-containing cup and tb contents 
.ixed several times with a pipette. After 
incubation, 100 ul of CaCl., prewarmed at 3^ C 
.o the plasma-actin reagent mixture using ^ -toma c 
Pipette/Timer-trigger <Becton-DicHinson. .he ^^^^^^ 

rcLrtinr:: r-tir orfi: i con e nations 

^-o^al assay volvune of 30U ux. 
of anticoagulant m the "tal as y ^^^^^ ,^ 

The nominal concentration of Factor ix 

"-^T^^-results shown in .ig. 1 -"-"--f/jr 
increasing concentrations of mu-e an i-.act^^ 

XX n^s BCl and BC2 on ^3 
inhibit clotting by prolonging the aPTT and b 

:each a final ^ -/^-aL^-sriTo: BCran^^C^ , 

rp::reir:r t:: drff:ence m the maximum response 

- - -^--rBrl Lrret^hrarTTabout to 

=r::rr ol Ther hand, increases the aPTT by 

; s-fold to about 90 sec. The therapeutic target zone 
.sed in anticoagulant therapy with -P^"; ^ 
highlighted. The results indicate that the 
bracket the heparin therapeutic aPTT range. 



P50438-1 



The properties of n^s BCl and BC2 are 

■ ^ in Table I Each of the BC mAbs recognizes 
summarxzed xn Table I ^^^.^^ 
both the zymogen, Factor IX, as well a 
protease, Factor IXa, but only BC2 .s -P^^^^; 
Locking both zymogen activation as well ^^ J^TZ^ 
activity. BCl and BC2 were found to -oss-react w 
Cynomologous monkey Factor IX. Additionally, BC2 also 
cross-reacted with rat Factor IX. 



Table I. 
IX mAbs 



sugary of in vitro Properties of ^nti-Factor 



BCl 



BC2 



Binds Factor IX 

Binds Factor IXa 

inhibits IX to IXa 
conversion 

Inhibits IXa activity 
in Xase complex 

Cofactor requirement 



yes 
yes 



yes 



yes 
yes 
yes 



divalent 
metals 



aPTTmax X 100% 
aPTTnormal 

ICso, 

Species cross- 
reactivity 



150 



-35 



Ca > Mn 
350 



monkey rat, monkey 
IgGl IgG2a 



39 



P50438-1 



The results shown in Fig. 2 demonstrate the effect 
of increasing concentrations of the anti-Factor IX mAbs 
9E4 (2) F4 and 11G4(1)B9 on aPTT clotting times. The 
plasma for the assay was diluted to one-half the normal 
concentration, giving an initial aPTT of 45 seconds. 
Both msbs inhibit clotting by prolonging the aPTT and 
both mAbs reach a final saturating effect on the aPTT. 
saturating concentrations of 9E4,2)F4 and 
increases the aPTT to -90 to 100 seconds for 9E4(2)F4 
and to -80 seconds for 11G4(1)E9. The results indicate 
that the two mAbs are at the upper end of the heparxn 
therapeutic aPTT range. 

The results shown in Fig. 3 demonstrate the effect 
of increasing concentrations of the anti-Factor X mAbs 
HFXLC (vs. light chain epitope). HFXHC (vs. heavy charn 
epitope) and the anti-Factor XI mab HFXI on aPTT 
clotting times. These :^s were obtained ^^^^ 
Research Laboratories (South Bend, IN) . The mAbs HFXLC 
and HFXI inhibit clotting by prolonging the aPTT and 
both mAbs reach a final saturating effect on the aPTT. 
The IC value for HFXLC is -40 nM; saturating 
concentrations increase the aPTT to -60 seconds. The 
IC value for HFXI is -20 nM; saturating concentratxons 
increase the aPTT to -100 seconds. The results indicate 
that HFXLC is within the heparin therapeutic aPTT range 
while HFXI falls at the upper end of the heparxn 
therapeutic range. The mAb HFXHC had no effect on aPTT 

clotting times. 

Self-limiting prolongation of the aPTT was also 
observed with antibodies to Factor VIII. the cof actor to 
Factor IXa. For example, the anti-human Factor VIII 
antibody. SAF8C-IG, purchased from Affinity Biologxcals, 
inc increased the aPTT to a maximum of about 65 sec. 
Half-maximal prolongation of the aPTT was achieved wrth 
about 100 nM antibody. 



40 



P50438-1 



Example 3 

' 7n Lder to ev aluate the efficacy of anti-Factor IK 
antibodies in prevention of arterial throinbosis, the rat 
carotid artery thrombosis model as reported by 
™cher et al. in . Car.ic ..ar., 526-533 (1393, 

was adapted. This model consists of segmental -3"-^ ^° 

the carotid endothelium by oxygen radicals generated by 

Feci, solution applied on the surface of the carotrd 

artery . 

in brief, rats were anesthetized with 
pentobarbitone sodium, the jugular vein cannulated for 
intravenous injections and the left femoral artery 
cannulated for blood pressure and heart rate monrtorxng. 
The carotid artery was isolated by aseptic technxque vxa 
a surgical incision in the neck and equipped wxth a 
magnetic flow probe for blood flow measurement. After a 
period of stabilization, baseline parameters were 
established for the following variables: carotid blood 
flow, arterial pressure, heart rate, activated partial 
thromboplastin time (aPTT) and prothrombin trme (PT)^ 
Thereafter, a premeasured Whatman filter paper soaked xn 
50% Feci, solution was placed on the carotid artery for 
15 minutes for complete injury of the underlyxng 
endothelial cells. After removal of the FeCl. soaked 
paper, the experiment was followed to completion over 
minutes. At the end of the experiment, the carotxd 
throTtous was extracted from the carotid artery and 

weighed. . 

All agents were administered 15 minutes prxor to 
the onset of carotid injury. The following treatments 
were examined and compared to the Factor IX mAb BC2 . 

1 Heparin: 15, 30, 60 or 120 U/kg bolus, 
followed by infusion of 0.5, 1, 2 or 4 U/kg/min, 
respectively over 60 minutes 



60 



41 



P50438-1 



2. Acetylsalicylic acid (ASA, aspirin): 5 mg/kg 

bolus , - 

3. Anti-Factor IX mAb BC2 : 1 , 3 or 6 mg/kg bolus, 

followed by infusion 0.3, 1, or 2 ug/kg/min, 
respectively over 60 minutes 

4. Heparin: 30U/kg bolus + lU/kg/min + ASA at 5 

Anti-Factor IX mAb BC2 : 1 mg/kg .0.3 ug/kg/min 
+ ASA at 5 mg/kg 

Figs 4 and 5 demonstrate the comparative 
pharmacology of the ant i -coagulant /thror^otic regimens 
by showing the effect of heparin, ASA and Factor IX mAb 
BC2 on aPTT (Fig. 4) and PT (Fig. 5). 

The key index for bleeding diathesis, aPTT, was 
used as the primary criterion for evaluation of efficacy 
versus bleeding liabilities of the anti- 
coagulant/ thrombotic agents used in the study. The 
results in Fig. 4 demonstrate the dose -dependent 
prolongation of aPTT by heparin with maximal 
prolongation of the clotting time, beyond the test 
limit, at the two higher doses. ASA alone dxd not 
significantly increase aPTT but in combination wxth 
heparin, a marked synergistic effect was observed. The 
Factor IX mAbs had a modest effect on aPTT and even at 
the highest dose, the increase in clotting time did not 
exceed the 3-fold limit of standard anti-coagulant 
practiced clinically. Most notably, the low dose of 
Factor IX mAb BC2 in combination with ASA did not change 
the aPTT. 

in Fig 5, the data indicate that PT was also 
significantly prolonged by heparin, at the two higher 
doses, and by the ASA . heparin combination, but not by 
any of the Factor IX mAb doses alone or in combination 
with ASA. 



42 



P50438-1 



The effect of heparin, ASA and Factor IX mAb on 
carotid artery occlusion is shown in Fig. 6. The 
results indicate that the carotid arteries of all of the 
vehicle-treated animals occlude in response to the 
injury. Heparin dose dependently inhibited the 
occlusion of the carotid artery. At the highest dose, 
heparin completely prevented the occlusion of the 
carotid artery; at this dose however, no coagulation 
could be initiated. ASA alone had only a minor effect 
on carotid occlusion. ASA in combination with heparin 
also failed to completely prevent carotid occlusion. 
Factor IX mAb completely blocked carotid occlusion at 
the two higher doses, which have not prolonged 
coagulation beyond the clinically desired target. The 
lower dose of Factor IX mAb, that largely failed to 
secure patency alone, demonstrated complete inhibition 
of carotid occlusion when administered in combination 
with ASA. 

The effect of heparin, ASA and Factor IX mAb on 
thrombus weight is shown in Fig. 7. Heparin dose- 
dependently reduced thrombus mass in the carotid 
artery. However, some residual thrombus was still found 
in the carotid artery in spite of complete blockade of 
coagulation. ASA alone or in combination with heparin 
(30 U/kg regimen) had only a partial effect on thrombus 
weight. Factor IX mAb dose-dependent ly reduced thrombus 
mass and the high dose virtually prevented completely 
thrombus formation. Moreover, the combination of the 
low dose anti-Factor IX mAb and ASA, a regimen that 
completely prevented carotid occlusion without adversely 
affecting the coagulation indices, completely prevented 
thrombus formation. 

The studies conducted in the rat carotid thrombosis 
model clearly demonstrate the efficacy of Factor IX mAb 
in prevention of thrombosis in a highly thrombogenic 
arterial injury model. Most notably, the efficacy of 



43 



P50438-1 



the Factor IX mAb was demonstrated within the desired 
therapeutic anticoagulant target defined by the aPTT. 
Furthermore, heparin, the current standard 
anticoagulant, reached efficacy comparable to Factor IX 
mAb only at doses that severely compromised coagulation 
to the extent of producing non-coagulable blood, 
interestingly, the observed potentiation and synergy 
acquired by ASA joint treatment with heparin was also 
demonstrated when ASA was given with anti-Factor IX mAb 
However, unlike the combination of heparin and ASA which 
resulted in potentiation of both the anti-thrombotic and 
anti-coagulant effects, the combination of Factor IX mAb 
and ASA resulted in potentiation of the anti-thrombotxc 
efficacy with no consistent effect on ex vivo blood 
coagulation parameters. Taken together, the data show a 
superior antithrombotic capacity of Factor IX mAb 
compared to heparin, ASA or a combination of heparin and 



ASA. 



Example 4 

^-.^.^^^r. Micr of^roPY of R at Thrombosis Model 

Segments of rat carotid artery were collected from 
sham, ferric chloride only and ferric chloride -h 6 mg/kg 
Factor IX antibody, 3/group, 15 minutes after 
application of ferric chloride. The arteries were fixed 
by perfusion with formaldehyde and ligated above and 
below the lesioned area. Fixed arteries were 
dehydrated, incubated in hexamethyldisilazane and dried 
in a desiccator. Dried arteries were opened lengthwise, 
placed on Scanning Electron Microscopy (SEM) stubs and 
sputter coated with gold. 

SEM of sham arteries revealed an essentially normal 
endothelium with rare scattered platelets. There were a 
few breaks in the endothelium, probably as a result of 
mechanical damage during surgery and the underlying 
basement membrane was covered by a carpet of platelets. 



44 



P50438-1 



NO evidence of thrombus formation was observed in the 

sham rats . , • j 

SEM of the arteries treated with ferric chloride 
revealed large mural thrombi which occupied a large 
portion of the lumen of the vessel. The thrombi were 
composed of aggregated platelets, red blood cells and 
amorphous and fibrillar proteinaceous material. The 
proteinaceous material is consistent with fibrin. The 
endothelium of the arteries was mostly obscured by the 
large thrombi, where visible, the endothelium overlyrng 
the region treated with ferric chloride was covered by 
numerous adherent platelets and amorphous proteinaceous 

material . . ■ j 

SEM of the arteries treated with ferric chlorxde 
from rats also treated with Factor IX antibody, revealed 
the lumen of the vessels to be largely free of thrombus. 
The endothelium overlying the region treated with ferrxc 
chloride showed extensive damage and some areas were 
covered by adherent platelets and platelet aggregates 
but there was little or no proteinaceous material. 

Example 5 
HtAb P^^ "-^^^ and T.ight-. Chain cDNA 
ff^rpiQTinft Analysis 

Total RNA was purified by using TriReagent 
(Molecular Research Center, Inc., Cincinnati, OH) 
according to the manufacturer's protocol. RNA was 
precipitated with isopropanol and dissolved in 0.5% SDS 
and adjusted to 0 . 5M NaCl . Poly A"^ RNA was isolated 
with Dynabeads Oligo (dT)25 (^nal A.S., Lake Success, 
NY) according to the manufacturer's protocol. Poly A 
RNA was eluted from the beads and resuspended xn TE 
buffer. Twelve aliquots of 100 ng of RNA were reverse 
transcribed with a RT-PCR kit per the manufacturer's 
instructions (Boehringer Mannheim Cat. No. 1483-188) 
using a dT oligo for priming. For the heavy chaxn, PCR 



45 



P50438-1 



amplifications of 6 RNA/DNA hybrids were carried out for 
25 cycles using a murine IgG2a hinge primer (SEQ ID NO: 

1) and a heavy chain signal sequence primer (SEQ ID NO: 

2) . Similarly, for the light chain, PGR amplif icatons 
of 6 RNA/DNA hybrids were carried out for 25 cycles 
using a murine kappa primer (SEQ ID NO: 3) and a 
degenerate light chain signal sequence primer (SEQ ID 
NO- 4) The PGR products from each of the 12 
amplifications were ligated in a PGR2000 vector (TA 
cloning Kit, Invitrogen, Cat. No. K2000-01) . Golonxes 
of recombinant clones were randomly picked and 
minipreparations of plasmid DNA were prepared using an 
alkaline extraction procedure described by Birnboim and 
Doly in Nucl. Acids Res. 1, 1513 (1979). The isolated 
plasmid DNA was digested with EcoRI and analyzed on a 

0 8% agarose gel. Double-stranded cDNA inserts of the 
appropriate size, i.e., -700 bp for the heavy chaxn and 
-700 bp for the light chain, were sequenced by a 
modification of the Sanger method. The sequence of all 
12 of the heavy and light chains were compared to 
generate a consensus BG2 heavy chain variable region 
sequence (SEQ ID NO: 5) and consensus BC2 light chain 
variable region sequence (SEQ ID NO: 6). 

sequence analysis of the BC2 heavy chain variable 
region cDNA revealed a 363 nucleotide open reading frame 
encoding a 121 amino acid sequence (SEQ ID NO: 7) . The 
heavy chain GDRl, 2 and 3 sequences are listed in SEQ ID 
NOs- 8, 9 and 10, respectively. 

sequence analysis of the BG2 light chain variable 
region cDNA revealed a 321 nucleotide open reading frame 
encoding a 107 amino acid sequence (SEQ ID NO: 11) • The 
light chain GDRl, 2 and 3 sequences are listed in SEQ ID 
NOs: 12, 13 and 14, respectively. 



46 



P50438-1 



Eacamole 6 
Tft,«.aTiigeti Antibodies 

Six humanized antibodies designated SB 249413, SB 
249415, SB 249416, SB249417, SB 257731 and SB 257732 
were designed to contain the murine CDRs described above 
in a human antibody framework. 
SB 249413 

SB 249413 contains the heavy chain F9HZHC 1-0 and 
the light chain F9HZLC 1-0. The synthetic variable 
region humanized heavy chain F9HZHC 1-0 was designed 
using the first three framework regions of the heavy 
chain obtained from immunoglobulin RF-TS3'CL (Capra, 
J D et al., J. Clin. Invest. 86, 1320-1328 (1990) 
identified in the Kabat database as Kabpro : HhclOw) and 
the BC2 heavy chain CDRs described previously. No 
framework amino acids substitutions which might 
influence CDR presentation were made. Four overlapping 
synthetic oligonucleotides were generated (SEQ ID NOs : 
15 16, 17 and 18) which, when annealed and extended, 
code for the amino acids representing the heavy chain 
variable region through and including CDR3 (SEQ ID NOs: 
19 and 20) . This synthetic gene was then amplified 
using PCR primers (SEQ ID NOs: 21 and 22) and ligated 
into the PCR2000 vector (TA cloning Kit, Invitrogen, 
cat. NO. K2000-01) and isolated from a Spel, Kpnl 
restriction digest. A second DNA fragment coding for 
the campath signal sequence including the first five 
amino acids of the variable region (SEQ ID NOs: 23 and 
24) was made by PCR amplification of the appropriate 
region of a construct encoding a humanized anti- 
Respiratory Syncitial Virus heavy chain (SEQ ID NO: 25) 
with two primers (SEQ ID NOs: 26 and 27) and digesting 
with the restriction enzymes EcoRI and Spel . The two 
fragments generated were ligated into an EcoRl, Kpnl 
digested pFHZHC2-6pCD mammalian cell expression vector 
which contained the remainder of a human consensus 



47 



P50438-1 



framework 4 and IgGl constant region. The vector 
contained a single amino acid mutation of the pFHZHC2- 
3pCD vector described in published International Patent 
Application NO. WO94/05690. The final residue of 
framework 2 (residue 49) was mutated from Ser to Ala by 
digesting pFHZHC2-3pCD with Xbal and EcoR5 and inserting 
a linker generated from two synthetic oligonucleotides 
(SEQ ID NOs: 28 and 29) . The sequence of the F9HZHC 1 0 
insert is shown in SEQ ID NOs: 3 0 and 31. 

The synthetic variable region humanized light chain 
F9HZLC 1-0 was designed using the framework regions of 
the human light chain obtained from immunoglobulin 
LS8'CL (Carmack et al . , J. Exp. Med. 169, 1631-1643 
(1989) identified in the Rabat database as 
Kabpro:Hkl318) and the BC2 light chain CDRs described 
previously. No framework amino acids substitutions 
which might influence CDR presentation were made. Two 
overlapping synthetic oligonucleotides were generated 
(SEQ ID NOs: 32 and 33) which, when annealed and 
extended, code for amino acids representing the light 
chain variable region (SEQ ID NOs: 34 and 35) . This 
synthetic gene was then amplified using PGR primers (SEQ 
ID NOs: 36 and 37) and ligated into the pCR2000 vector 
(TA cloning Kit, Invitrogen, Cat. No. K2000-01) , and 
isolated from a Seal, 5acII restriction digest. A 
second DNA fragment coding for the campath signal 
sequence including the first two amino acids of the 
variable region (SEQ ID NOs: 38 and 39) was made by PGR 
amplification of the the appropriate region of a 
construct encoding a humanized anti-Respiratory 
Syncitial Virus heavy chain (SEQ ID NO: 25) with the two 
primers (SEQ ID NOs: 26 and 40) and digesting with the 
restriction enzymes EcoRl and Seal. The two fragments 
generated were ligated into an EcoRl, SacII digested 
pFHzLGl-2pCN mammalian cell expression vector which 
contained the remainder of a human framework 4 and kappa 



48 



P50438-1 



constant region. The vector contained a single amxno 
acid mutation of the pFHZLCl-lpCN vector described xn 
published international Patent Application No. 
WO94/05690. A framework 2 residue was mutated from Ser 
to Pro by digesting pFHZLCl-pCN with Smal and Kpnl and 
inserting a linker generated from two synthetic 
oligonucleotides (SEQ ID NOs: 41 and 42). The sequence 
of the F9HZLC 1-0 insert is shown in SEQ ID NOs: 43 and 



44. 



SB 249415 ^ ^ 

SB 249415 contains the heavy chain F9HZHC 1-1 and 
the light chain F9HZLC 1-1- These heavy and light chain 
constructs are based on F9HZHC 1-0 and F9HZLC 1-0, 
respectively, however, they have framework amino acid 
substitutions which can influence CDR presentation. 

F9HZHC 1-1 has three framework amino acid 
substitutions which might influence CDR presentation. 
TWO overlapping synthetic oligonucleotides were 
generated (SEQ ID NOs: 45 and 46), which when annealed 
and extended, code for amino acids representing the 
altered portion of the heavy chain variable region 
altered (SEQ ID NOs: 47 and 48). This synthetic gene 
was then amplified using PGR primers (SEQ ID NOs: 49 and 
50), ligated into the pCR2000 vector (TA cloning Kit, 
invitrogen, Cat. No. K2000-01) and isolated from a 
EcoNI, Kpnl restriction digest. This fragment was 
ligated into EcoNI, Xpnl digested F9HZHC1-0 (SEQ ID NO: 
30) vector. The sequence of the F9HZHC 1-1 insert is 
shown in SEQ ID NOs: 51 and 52. 

F9HZLC 1-1 has four framework amino acids 
substitutions which can influence CDR presentation. TVo 
synthetic oligonucleotides were generated (SEQ ID NOs: 
53 and 54), which when annealed, have Kpnl and BaiiTHI 
cohesive ends, and code for amino acids representing the 
altered portion of the light chain variable region (SEQ 
ID NO: 55). F9HZLC 1-0 (SEQ ID NO: 43) was digested 



49 



P50438-1 



with the restriction enzymes Kpnl and BaMll and ligated 
to the synthetic DNA. The sequence of the F9HZLC 1-1 
insert is shovm in SEQ ID NOs : 56 and 57. 
SB 249416 

SB 249416 contains the heavy chain F9HZHC 1-1 
(described above) (SEQ ID NO: 52) and the light chain 
F9HZLC 1-2. The light chain construct is based on 
F9HZLC 1-1, however, it has one additional framework 
amino acid substitution which can influence CDR 

presentation. 

TWO synthetic oligonucleotides were generated (SEQ 
ID NOs: 58 and 59), which when annealed, have BamHI and 
Xbal cohesive ends and code for amino acids representxng 
the altered portion of the light chain variable regxon 

(SEQ ID NO: 60) . F9HZLC 1-1 (SEQ ID NO: 56) vector was 
digested with the restriction enzymes BaMll and Xbal and 

ligated to the synthetic DNA. The sequence of the 

F9HZLC 1-2 insert is shown in SEQ ID NOs: 61 and 62. 

.qg 249417 

SB 249417 contains the heavy chain F9HZHC 1-1 
(described above) (SEQ ID NO: 52) and the light chain 
F9HZLC 2-0. A F9HZLC 2-0 synthetic variable region 
humanized light chain was designed using the framework 
regions of the human light chain obtained from 
immunoglobulin REI (Palm and Hilschmann, Z. Physiol. 
Chem 354. 1651-1654 (1973) identified in the Kabat 
database as Kabpro : HKLlll) and the BC2 light chain CDRs 
described previously. Five amino acid consensus human 
substitutions were introduced. Six framework amino 
acids murine substitutions which can influence CDR 
presentation were made. Two overlapping synthetic 
oligonucleotides were generated (SEQ ID NOs: 63 and 64) 
Which, when annealed and extended, code for amino acids 
representing the light chain variable region (SEQ ID 
NOS- 65 and 66) . This synthetic gene was then amplified 
using PGR primers (SEQ ID NOs: 67 and 68), ligated into 



50 



P50438-1 



the PCR2000 vector (TA cloning Kit, Invitrogen, Cat. No. 
K2000-01) and isolated from a Seal, SacII rastrictxon 
digest A second DNA fragment coding for the campath 
signal sequence including the first two amino acids of 
the variable region (SEQ ID NO: 38) was made by PCR 
amplification of the the appropriate region of a 
construct encoding a humanized anti-Respiratory 
syncitial Virus heavy chain (SEQ ID NO: 25) with two 
primers (SEQ ID NOs : 26 and 69) and digesting wrth the 
restriction enzymes EcoRI and Seal. A third DNA 
fragment encoding the remainder of a human framework 4 
(SEQ ID NO: 70) and having SacII and Narl cohesive ends 
was generated by annealing two synthetic 
oligonucleotides (SEQ ID NOs: 71 and 72). F9HZLC 1-0 
(SEQ ID NO: 43) was digested with the restriction 
enzymes EcoRI and Narl and ligated to the three DNA 
fragments. The sequence of the F9HZLC 2-0 insert is 
shown in SEQ ID NOs: 73 and 74. 
SB 257731 

iri57731 contains the heavy chain F9HZHC 1-1 (SEQ 

ID NO: 52) and the light chain F9HZLC 1-3, a single 
axnino acid mutation of F9HZLC 1-2 (SEQ ID NO: 62). 
F9HZLC 1-2 was PCR amplified with two primers (SEQ ID 
NOS- 26 and 69) and digested with the restriction 
enzymes EcoRI and Seal. A 94 bp fragment (SEQ ID NOs: 
75 and 76) was isolated. The fragment was Ixgated xnto 
EcoRI, seal digested F9HZLC 1-2 vector to produce the 
light chain construct F9HZLC 1-3. The sequence of the 
F9HZLC 1-3 insert is shown in SEQ ID NOs: 77 and 78. 
SB 257732 

ir^57732 contains the synthetic variable regxon 
humanized heavy chain F9HZHC 3-0 and light chain F9HZLC 
3-0 Four overlapping synthetic oligonucleotides were 
generated (SEQ ID NOs: 79, 80, 81 and 82) which, when 
annealed and extended, code for the amino acids 
representing the heavy chain variable region being 



51 



P50438-1 



altered (SEQ ID NOs: 83 and 84). This synthetic gene 
was then amplified using PGR primers (SEQ ID NOs: 85 and 
86) ligated into the pCR2000 vector (TA cloning Kit, 
invitrogen. Cat. No. K2000-01) and isolated from a 5tul, 
Kpnl restriction digest. The isolated fragment was 
ligated into Stul , Kpnl digested F9HZHC1-1 (SEQ ID NO: 
52) vector. This vector was then digested with EcoRI, 
Spel to remove the signal sequence. A DNA fragment 
coding for the campath signal sequence (SEQ ID NO: 23) 
including the first five amino acids of the variable 
region was made by PGR amplification of F9HZHG1-0 with 
two primers (SEQ ID NOs: 2 6 and 87) and digesting wxth 
the restriction enzymes EcoRI and Spel . The fragment 
generated was ligated into the vector. The sequence of 
the F9HZHG3-0 insert is shown in SEQ ID NOs: 88 and 89. 

Four overlapping synthetic oligonucleotides were 
generated (SEQ ID NOs: 90, 91, 92 and 93) which, when 
annealed and extended, code for amino acids representing 
the light chain variable region (SEQ ID NOs: 94 and 95). 
This synthetic gene was then amplified using PGR primers 
(SEQ ID NOs: 96 and 97) and ligated into the pGR2000 
vector (TA cloning Kit, Invitrogen, Gat. No. K2000-01) , 
and isolated from a 5cal, I^arl restriction digest. The 
isolated fragment was ligated into Seal, Narl digested 
F9HZLC1-3 (SEQ ID NO: 77) vector. The sequence of the 
F9HZLC3-0 insert is shown in SEQ ID NOs: 98 and 99. 

The humanized anti-Factor IX mAbs were expressed xn 
CHO cells. A DG-44 cell line adapted for suspension 
growth in serum-free medium was grown in 100ml of 
protein-free medium containing IX nucleosides and 0.05% 
F68 in 250 ml disposable sterile erlenmeyer flasks 
(Corning) on a Innova 2100 platform shaker (New 
Brunswick Scientific) at 150 rpm at 37°C in a 5% GO,, 95% 
air humidified incubator. These cells were passaged at 
4 X 10' cells/ml twice weekly. 15 ug each of the pCN-Lc- 
Light Chain and pGD-Hc-heavy chain vectors were 



52 



P50438-1 



linearized by digestion with i^otl, co-precipitated under 
sterile conditions and resuspended in 50ul of IX TE 
buffer (lOmM Tris, linM EDTA, pH 7.5) . The DNA was 
electroporated using a Bio-Rad Gene Pulser (Bio-Rad 
Laboratories) into the Acc-098 cells using the technique 
of Hensley at al . in J. Biol. Chem. 269. 23949-23958 
(1994) . 1.2 X 10' cells were washed once in 12.5 ml of 
ice cold PBSucrose (PBS, 272mM sucrose, 7mM sodium 
phosphate pH 7.4, ImM MgCl,) , resuspended in 0.8 ml of 
PBS added to 50ul of the DNA solution and incubated on 
ice for 15 min. The cells were pulsed at 380 V and 25 
microfarads, then incubated on ice for 10 min. Cells 
were plated into 96 well culture plates at 5 X 10 
cells/plate in maintenance medium for 24 hr prior to 
selection. Cells were selected for resistance to 
400ug/ml G418 (Geneticin, Life Technologies, Inc.) in 
maintenance medium. 24 hr prior to assay, the cells 
were fed with 150ul of the maintenance medium. 

conditioned medium from individual colonies was 
assayed using an electrochemiluminescence (ECL) 
detection method on an Origen analyzer (IGEN, Inc . ) . 
5ee Yang et al . , Biotechnology, 12. 193-194 (1994). 

All solutions necessary for the performance of the 
assays {assay buffer) and for the operation of the 
analyzer (cell cleaner) were obtained from IGEN. The 
antibodies (anti-human IgG (g-chain specific) , Sigma 
Chemicals and F(ab')3 Fragment to Human IgG (H+L) , 
Kirkegaard & Perry Laboratories Inc.) were labelled with 
TAG-NHS-ester (IGEN, Inc.) at a 7 : 1 molar ratio of 
TAG:protein, while the Protein A (Sigma) was labelled 
with Biotin-LC-Sulfo-NHS-ester (IGEN, Inc.) at a 20:1 
molar ratio Biotin :protein, both according to IGEN's 
recommendations. Streptavidin-coated magnetic beads (M- 
280) were obtained from Dynal . 

immunoassays were performed using the following 
protocol: per sample, 50ul of the streptavidin-coated 



53 



P50438-1 



beads (final concentration 600 ug/ml diluted in PBS, 
pH7.8, with 1.25% Tween) were mixed with 50ul Biotin- 
Protein A (final concentration lug/diluted in PBS, 
pH7.8, with 1.25% Tween) and incubated at room 
temperature for 15min with agitation, 50ul of the TAG 
antibodies (a mixture with a final concentration of 1.25 
ug/ml F(ab'), Fragment to Human IgG (H+L) and 0.25 ug/ml 
Anti-Human IgG (g-chain specific) diluted in PBS, pH7.8, 
with 1.25% Tween) were added, the solution was then 
added to 50ul of conditioned medium and incubated with 
agitation at room temperature for 1 hr. 200ul of assay 
buffer was added to the reaction mix and the sample 
analyzed on the Origen I analyzer to measure ECL. The 
results indicated that approximately 20-37% of the 
colonies assayed secrete over 15 ng/ml of the antibody 
with an average expression of about 150 ng/ml. 

Humanized anti-Factor IX mAbs were purified from 
the conditioned media using a Procep A capture step 
followed by ion-exchange chromatography to reduce the 
DNA burden. Procep A sorbent material (Bioprocessing 
Ltd., Durham, England) was used to prepare a column with 
a 1:1 diameter to height ratio. Clarified conditioned 
media was loaded onto the column at about 150 cm/hr. 
The column was washed sequentially with phosphate 
buffered saline (PBS), PBS containing 1 M NaCl, and 
finally with PBS. The bound material was recovered with 
0.1 M acetic acid elution. The eluate was adjusted to 
pH 5.5 and was diluted (1:4) with water. The diluted 
solution was loaded onto an S-Sepharose column (2.5 x 13 
cm) which was pre-equilibrated with 2 0 mM sodium 
acetate, pH 5.5 at 80 cm/hr. The column was washed with 
the acetate buffer until a steady baseline was obtained 
and the bound protein was eluted with 20 mM sodium 
phosphate, pH 7.4 at 25 cm/hr. The eluted material was 
filtered with a 0.4 micron membrane and stored at 4^0. 



54 



P50438-1 



Example 7 
Mouae-Hum&n Chimer ic Antibody 

100 ng of BC2 RNA were reverse transcribed with a 
RT-PCR kit per the manufacturer's instructions 
(Boehringer Mannheim Cat. No. 1483-188) using a dT oligo 
for priming, and PGR amplified with synthetic Seal (SEQ 
ID NO: 100) and Narl {SEQ ID NO: 101) primers to produce 
the BC2 light chain variable region with Seal, Narl ends 
(SEQ ID NOs: 102 and 103) . This DNA was ligated into 
Seal, Warl digested F9HZHC1-3 (SEQ ID 77) and digested 
with Seal, Narl to produce a mouse-human chimeric light 
chain F9CHLC (SEQ ID NOs: 104 and 105) . 

100 ng of BC2 RNA were reverse transcribed with a 
RT-PCR kit per the manufacturer's instructions 
(Boehringer Mannheim Cat. No. 1483-188) using a dT oligo 
for priming, and PCR amplified with synthetic Spel (SEQ 
ID NO: 106) and Nhel (SEQ ID NO: 107) primers to produce 
the BC2 heavy chain variable region with Spel, Nhel ends 
(SEQ ID NOs: 108 and 109). The campath signal sequence 
was PCR amplified from the RSVHZ19 heavy chain (SEQ ID 
NO: 25) with EcoRI (SEQ ID 26) and Spel (SEQ ID 87) 
primers. These two DNA fragments were ligated into a 
BcoRI, Nhel digested lL4CHHCpcd vector described in 
published International Patent Application No. 
WO95/07301, replacing the IL4 variable region with the 
BC2 Factor IX mouse variable region, to produce a mouse- 
human chimeric heavy chain F9CHHC (SEQ ID Nos : 110 and 
111) - 

Co-transfection and purification of the mouse-human 
chimeric antibody chaFIX was accomplished as described 
above for the humanized constructs. 



55 



P50438-1 



Exam.pl 8 

m^r^rj^^^Y >i»ni« «^ Facto ^ IX mAbs in Rat Thrombus 
Model 

In order to evaluate the efficacy of humanized 
anti-Factor IX antibodies in prevention of arterial 
thrombosis, the rat carotid artery thrombosis model as 
described above in Example 3 was used. Baseline 
parameters were established for carotid blood flow, 
arterial pressure, heart rate, vessel patency and 
activated partial thromboplastin time (aPTT) . Fifteen 
minutes thereafter, carotid injury was effected for 10 
minutes. The parameters were determined 60 minutes 
after onset of carotid injury. Carotid thrombus was 
also extracted from the carotid artery and weighed. 

All agents were administered intravenously 15 
minutes before the onset of carotid injury. The 
following treatments were examined and compared to the 
anti-Factor IX mAb BC2 . 

1 . Vehicle 

2. chaFIX: 3 mg/kg bolus 

3. SB 249413: 3 mg/kg bolus 

4. SB 249415: 3 mg/kg bolus 

5. SB 249416: 3 mg/kg bolus 

6. SB 249417: 3 mg/kg bolus 

7. SB 257731: 3 mg/kg bolus 

8. Heparin: 60 units /kg bolus + 2 units /kg/min 
infusion 

The aPTT was used as the primary criterion for 
evaluation of efficacy versus bleeding liabilities of 
the anti-coagulant/ thrombotic agents used in the study. 
The results in Fig. 8 demonstrate that the humanized 
Factor IX mAbs SB 249413, SB 249415, SB 249416, SB 
249417 and SB 257731 had a modest effect on aPTT at 3 . 0 
mg/kg which is within the clinical accepted range. 



56 



P50438-1 



The effect of the Factor IX mAbs on thrombus mass 
is shown in Fig. 9. The results indicate that all of 
the humanized mAbs are equally effective in reducing 
thrombus mass . 

The studies conducted in the rat carotid thrombosis 
model clearly demonstrate the efficacy of the humanized 
Factor IX mAbs in prevention of thrombosis in a highly 
thrombogenic arterial injury model. Most notably, the 
efficacy of all of the humanized Factor IX mAbs was 
demonstrated within the desired therapeutic 
anticoagulant target defined by the aPTT. 

Exeunple 9 

Antibody Biochemical and B iophysical Properties 

The molecular mass of SB 249417 was determined by 
MALD-MS to be 148,000Da. Analytical ultracentrif ugation 
of SB 249417 gave an identical value. In the presence 
of Factor IX plus Ca2+, the antibodies derived from BC 2 
sedimented with a mass of 248,000Da corresponding to the 
combined mass of the mAb and two molecules of Factor IX. 
No evidence of higher ordered aggregates was observed in 
the presence or absence of Factor IX. 

The kinetics of Factor IX binding to SB 249417 was 
assessed by BIAcore analysis with antibody bound to an 
immobilized protein A surface. Recombinant human Factor 
IX (rhFIX, Genetics Institute) at 49 nM was used and 
measurements performed in the presence of 5 mM Ca2+. 
The interaction was characterized by rapid association, 
kass = 2.0 X 105 M"! s"! and relatively slow off-rate, 
kdiss = 4.1 X 10-4 s-1. The calculated for Factor IX 
binding was 1.9 nM. 

Table 1 summarizes the biophysical properties of SB 
249417 . 



57 



P50438-1 



Table 1 

Summary of the Biophysical Properties of SB 249417 

Isotype IgGl, kappa 

Purity by SDS-PAGE >95% (under reducing 

conditions) 

Molecular Weight 

Mass Spectrometry 148,000 Da 

Analytical Ultracentrifugation 148,000 Da 

Stoichiometry of Factor IX Binding 

Isothermal Titration Calorimetry 1.5 moles Factor 
IX: 1 mole mAb 

Factor IX Binding Affinity 

Isothermal Titration Calorimetry Kd= 4 nM at 25°C 
Biosensor Kd= 2 nM 

Factor IX Binding Kinetics 

Biosensor k^ss = 2.0 x 10^ M"! s'l 

kdiss =4 X 10-4 s-1 



Table 2 summarizes the factor IX binding properties 
of mAbs of the present invention. The calculated 
dissociation constants were essentially identical within 
experimental error. 



P50438-1 



Table 2 

Kinetics of Factor IX Binding to Anti-Factor IX mAbs 





249417 








calc 


. Kp <nM) 


SB 


2 . 


0 xl05 


4.1x10-4 




1 . 9 




BC2 


4 


.SxlO^ 


9. 


1x10-4 




1 . 9 




Chf9 


2. 


4 xl05 


3 . 


.0x10-4 




1 . 3 


SB 


249413 


6 . 


5 xl05 


2 . 


.8x10-3 


3 . 


.7-5.1 


SB 


249415 


7 . 


5 xl05 


1, 


.8x10-4 


1 


.1-2.3 


SB 


249416 


5. 


.2 xl05 


4 


.1x10-4 




0.8 


SB 


257731 


9 . 


,2 xl05 


9 


.9x10-4 




1.1 


SB 


257732 


1, 


.1 xl06 


1 


.2x10-3 




1.5 



The interactions between rhFIX and SB 249417 , BC2 
and other hiimanized constructs were characterized by 
titration microcalorimetry, which measures binding 
interactions in solution from the intrinsic heat of 
binding. Nine injections of 106 uM FIX were made into 
the calorimeter containing 2 uM. mAb SB 249417. Binding 
was detected in the first 4 injections as exothermic 
heats. At the last 5 injections the mAb binding sites 
were saturated with FIX and only background heats of 
mixing were observed. The results indicated that the 
equivalence point occurred at a molar binding ratio near 
2 FIX per mAb, as expected. Nonlinear least squares 
analysis of the data yield the binding affinity. 

The rhFIX affinities of the mAbs were measured over 
a range of temperature from 3 4-44°C in lOmM HEPES, lOmM 
CaCl2, 150mM NaCl, pH 7.4. These data allow the 
affinity at 37°C to be determined directly and the 
affinity at 25°C to be calculated from the van't Hoff 



59 



P50438-1 



equation. The data in Table 3 indicate that the 
affinities of SB 249417, BC2 and its other humanized 
constructs are within error (a factor of 2) the same. 

Table 3 

Titration Calorimetry Results for Anti-PIX loAbs 



Kd, nM at 25°C Kd, nM at 37°C 



Molar Binding 
Ratio 



BC2 

SB 
249413 

SB 
249415 

SB 
249417 

SB 
257732 



20 
12 



1.4 
1.9 



The mAbs SB 249413, SB 249415, SB 249417 and SB 
257732 all exhibited very similar thermal stabilities by 
differential scanning calorimetry. Their unfolding Tms 
ranged from 70-75°C indicating high stability against 
thermally induced denaturation . 



Example 10 

Mechanism of Antibody-Mediated I nhibition of Factor IX 

A library of chimeric constructs composed of 
sequences of Factor IX spliced into the framework of the 
homologous protein Factor VII was constructed and used 
to map the epitope for the Factor IX BC2 mAb. See 
Cheung et al . , Thromb. Res. 80, 419-427 (1995). Binding 
was measured using a BiaCore 2000 surface plasmon 



60 



P50438-1 



resonance device. The BC2 antibody was coupled directly 
to the chip using the NHS/EDC reaction. Binding was 
measured by 2 min of contact time at 20uL/min with 200 
nM of each of the given constructs in 25 mM MOPS, pH 
7.4, 0.15 M NaCl, 5 mM CaCl2 • Dissociation was 
monitored for 3 min using the same buffer with no 
protein. No binding was detected to the wild type 
construct in the presence of 50 mM EDTA. The data are 
presented in Table 4 . 

Table 4 

Summmary of Binding of Factor IX Constructs to BC2 
Antibody 





Construct 


Degree of Binding 




Plasma IXa 


Binds 




r-IX 


Binds 




Plasma VII 


No Binding 




IX LC/VII HC 


Binds 




IX-A/VII 


Binds 




VII gla/IX 


No Binding 




VII-A/IX 


No Binding 


VII 


gla {IX 3-11) /IX 


Binds 


VII 


gla (IX 3-6) /IX 


Very Low Binding 


VII 


gla (IX 9-11) /IX 


Very Low Binding 




IX K5A 


Binds 



These data indicate that the constructs containing 
the Factor IX light chain and Factor VII heavy chain (IX 
LC/VII HC) ; the Factor IX gla and aromatic stack domains 
(IX-A/VII); residues 3-11 of Factor IX gla domain within 



61 



P50438-1 



the Factor VII gla domain (VII gla (IX 3-11) /IX) ; and 
Factor IX having a lysine to alanine substitution at 
residue 5 (IX K5A) exhibit binding to BC2 . The VII gla 
{IX 3-11) /IX construct exhibited BC2 binding equivalent 
to wild type Factor IX (plasma IXa and r-IX) . Thus, the 
BC2 antibody binds to an epitope contained within 
residues 3-11 of the Factor IX gla domain. 

Example 11 

ni^^^i-m^TYt of Arte r n^l Thrombosis with Anti-Factor IX 
Antibody and Tisgue Plasm inogen Activator 

Administration of tPA with or without adjunctive 
therapies, was initiated following complete occlusion of 
the carotid artery. Blood flow in the artery was 
continuously monitored. 

Male Sprague-Dawley rats (Charles River, Raleigh, NC) 
weighing 3 00-490 gm were anesthetized with sodium 
pentobarbital (55 mg/kg, i.p.). The rats were placed 
dorsal on a heated (37°C) surgical board and an incision 
was made in the neck; the trachea was isolated and 
cannulated with a PE-240, Intramedic tube. The left 
carotid artery and jugular vein were then isolated. A 
Parafilm M sheet (4 mm2 , American National Can) was 
placed under the carotid artery and an electromagnetic 
blood flow probe (Carolina Medical) was placed on the 
artery to measure blood flow. A cannula (Tygon, 0.02" x 
0.04", Norton Performance Plastics) was inserted into 
the jugular vein for drug administration. The left 
femoral artery was then isolated and cannulated for 
measurement of blood pressure and collection of blood 
samples . 

Thrombosis in the carotid artery was initiated with 
a 6.5 mm diameter circular patch of glass micro-filter 
paper saturated with FeCls solution (50%) placed on the 
carotid artery downstream from the flow probe for 10 
minutes as described in Example 3 . In this well- 

62 



P50438-1 



characterized model, thrombus formation is usually 
complete within 15 min. 

The anti-Factor IX antibody, SB 249415 was 
administered as a bolus in combination with tPA 
(Genentech, South San Francisco, CA) , while heparin 
(Elkins-Sinn Inc., Cherry Hill, NJ)was administered as a 
bolus followed by infusion. All drug infusions 
continued to the end of the experimental period - 60 
minutes from the time of vessel occlusion. Blood 
samples, 1 mL, were collected for aPTT and FT assay at 
0, 30 and 60 min (end of study) from the femoral artery 
into 3.8% citrate solution and centrifuged. aPTT and 
prothrombin time (PT) were monitored by a fibrometer 
(BBIL, Baxter Dade or MLA Electra 800 Automatic 
coagulation Timer) with standard procedures. At the end 
of the experiment, the thrombus was extracted from the 
carotid artery and weighed. 

All data are presented as mean group values + SEM 
for the indicated number of rats in each group. ANOVA 
and Bonferoni tests for multiple comparisons were used 
for between group analyses and a p < 0.05 accepted as 
significance. 

Formation of an occlusive thrombus occurs 
approximately 15 min after initiation of arterial injury 
by application of the FeCls treated patch to the rat 
carotid artery. As shown in Fig. 10, with tPA alone, 
reperfusion of the occluded vessel was only observed 
following administration of a dose of 9 mg/kg tPA with 
67% of the treated vessels exhibiting regain of blood 
flow during the 60 min protocol. At this dose of tPA 
inclusion of 60 U/kg heparin or 3 mg/kg anti-Factor IX 
antibody, SB 249415, did not result in a further 
increase in the incidence of reperfusion suggesting that 
in the FeCl3 injury model about 30% of the thrombi are 
refractory to lysis. 



63 



P50438-1 



The results in Fig. 10 indicate that, at lower 
doses of tPA, the incidence of reperfusion is 
significantly dependent upon which anticoagulant was co- 
administered with the thrombolytic. When 60 U/kg 
heparin was administered the percentage of vessels 
showing reperfusion decreased dramatically with only 
12.5% and 40% reperfusion observed with 3 and 6 mg/kg 
tPA, repectively. Co-administration of 3 mg/kg SB 
249415 with the tPA, however, achieved greater than 60% 
reperfusion with 3 mg/kg tPA and 79% reperfusion with 
the 6 mg/kg tPA dose. Thus, the anti-Factor IX antibody 
significantly shifts the thrombolytic dose response 
curve allowing reperfusion with lower doses of 
thrombolytic agent. 

Thrombolysis and clot formation are dynamic 
processes and periods of patency followed by re- 
occlusion were sometimes observed. Since carotid blood 
flow was monitored continuously during the 60 min 
experimetal protocol, it was also possible to quantitate 
the total time of carotid patency. As shown in Fig. 11, 
the total period of vessel patency is substantially 
increased by combination of 3 mg/kg anti-Factor IX 
antibody plus tPA. This is particularly evident at the 
lowest and the intermediate doses of tPA, 3 and 6 mg/kg, 
respectively. At a combined dose of 3 mg/kg SB 249415 
plus 3 mg/kg tPA, the total patency time was 30.6 ± 9.2 
min compared to 7 . 1 ± 7 . 1 min f or the combination of 60 
U/kg heparin plus 3 mg/kg tPA. Patency time was zero 
with 3 mg/kg tPA alone. With a dose of 6 mg/kg tPA, co- 
administration with heparin increases patency time only 
slightly to 12.9 ± 6.0 min whereas the tPA-SB 249415 
combination achieves maximal patency time of 3 8.7 ± 8.4 
min. Only at the highest dose of tPA (9 mg/kg) does the 
heparin combination approach the patency achieved with 
SB 249415, 31.9 ± 4.8 min and 38.0 ± 8.4 min, 
respectively . 



64 



P50438-1 



Rapid restoration of blood flow following arterial 
infarct is critical to minimizing damage to the ischemic 
tissue. The results in Fig. 12 indicate that the 
combination of anti-factor IX antibody with tPA resulted 
in decreased time to reperfusion compared to tPA alone 
or heparin plus tPA and that this is achieved with lower 
doses of tPA. When thrombolysis was effected with 3 
mg/kg SB 249415 plus 3 mg/kg tPA the time to 
thrombolysis was 29.4 ± 9.2 min. With 3 mg/kg tPA, 
alone, no reperfusion was observed. With 60 U/kg 
heparin plus 3 mg/kg tPA the time to thrombolysis was 
52.8 ± 7.1 min. At higher doses of tPA, 6 and 9 mg/kg, 
the antibody plus tPA treatment regimen achieved initial 
thrombolysis in 19.4 ± 6.3 and 20.8 ± 8.7 min, 
respectively. In the absence of added anticoagulant, 
the time to thrombolysis was 60 min ( i.e., the limit of 
the experimental protocol) and 27.5 ± 6.4 min for doses 
of 6 and 9 mg/kg, respectively. With addition of 60 
U/kg heparin, the corresponding times to thrombolysis 
were 44.0 ± 7.1 and 27.0 ± 4.9 min. Thus, earlier 
reperfusion was always achieved with SB 249415 than with 
heparin or with tPA alone. 

Eagamole 12 

Effect of AT^ti-Facto r- TX Antibody on Hemosf.atlc Function 

The impact of anti-factor IX or heparin as 
adjunctive agents on the maintanence of hemostatic 
function was determined by monitoring levels of 
fibrinogen, plasminogen and alpha-2-antiplasmin at the 
end of the treatment period in rats treated with tPA 
alone, tPA plus heparin and tPA plus SB 249415 and the 
results were compared to vehicle treated animals. As 
shown in Fig. 13, increasing doses of tPA resulted in 
decreased levels of each of the hemostatic markers 
measured. Alpha-2-antiplasmin levels dropped from about 
90% in animals not treated with tPA to about 20% as the 



65 



P50438-1 



dose of tPA was increased to 9 mg/kg. Plasminogen 
levels dropped from an average of about 100% without tPA 
treatment to about 40% in the 9 mg/kg treatment group. 
Likewise, fibrinogen levels dropped from about 150 mg/dL 
to about 90 mg/dL in the high-dose tPA group, 
interestingly, the selection of the adjunctive agent 
does not appear to significantly effect any of these 
markers. At each tPA dose, similar levels of alpha-2- 
antiplasmin, plasminogen and fibrinogen were observed xn 
animals given vehicle, 30 or 60 U/kg heparin or 1 or 3 ^ 
mg/kg SB 249415; the observed decrease in the hemostatic 
markers is only a function of dose of the thrombolytic 
agent, tPA, and these decreases, especially in the case 
of fibrinogen, appear to be particularly large with tPA 
doses greater than 6 mg/kg, i.e., in the 9 mg/kg hxgh- 
dose group. 

The effects of the different treatment regimens on 
the standard aPTT coagulation assay aPTT was also 
monitored (Fig. 14). With increasing doses of tPA the 
aPTT increased from 19.3s ± 0.6s to 30.0s ±1.6s for 
vehicle and 9 mg/kg tPA, respectively. Administration 
of 3 mg/kg SB 249415 produced a limited increase m the 
aPTT of control animals to 49 . 6 s ± 6 . 4 s . When SB 
249415 was co-administered with tPA the observed 
increase was slightly larger and was dependent upon the 
dose of tPA. combination of SB 249415 with 3 mg/kg tPA 
produced an aPTT of 58.3 s ±5.2 s whereas combination of 
SB 249415 with the 9 mg/kg dose of tPA increased the 
aPTT to 77.3 s ± 19.7 s. Administration of either the 
3 0 U/kg or 60 U/kg dose of heparin resulted in large 
increases in the aPTT. Without tPA the aPTT ranged from 
about 300 s to 600 s for 30 and 60 U/kg doses, 
respectively. In tPA treated animals the aPTT was about 
800 s. 

The elevation of the aPTT obtained with heparin, 
particularly when coupled with the perturbation of 



66 



P50438-1 



hemostatic parameters due to the need for high doses of 
tPA to achieve effective reperfusion, is likely to 
contribute to bleeding liabilities. Conversely, SB 
249415 does not cause major elevation of the aPTT and 
enables the use of lower doses of tPA providing 
significant advantage in thrombolytic therapy in 
myocardial infarction and stroke. 

The present invention may be embodied in other 
specific forms without departing from the spirit or 
essential attributes thereof, and, accordingly, 
reference should be made to the appended claims, rather 
than to the foregoing specification, as indicating the 
scope of the invention. 



67